Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Neuroendocrinology ; 108(3): 172-189, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30537700

RESUMO

BACKGROUND/AIMS: The medial amygdala (MeA) responds to olfactory stimuli and alters reproductive physiology. However, the neuronal circuit that relays signals from the MeA to the reproductive axis remains poorly defined. This study aimed to test whether MeA kisspeptin (MeAKiss) neurons in male mice are sensitive to sexually relevant olfactory stimuli and transmit signals to alter reproductive physiology. We also investigated whether MeAKiss neurons have the capacity to elaborate glutamate and GABA neurotransmitters and potentially contribute to reproductive axis regulation. METHODS: Using female urine as a pheromone stimulus, MeAKiss neuronal activity was analysed and serum luteinizing hormone (LH) was measured in male mice. Next, using a chemogenetic approach, MeAKiss neurons were bi-directionally modulated to measure the effect on serum LH and evaluate the activation of the preoptic area. Lastly, using in situ hybridization, we identified the proportion of MeAKiss neurons that express markers for GABAergic (Vgat) and glutamatergic (Vglut2) neurotransmission. RESULTS: Male mice exposed to female urine showed a two-fold increase in the number of c-Fos-positive MeAKiss neurons concomitant with raised LH. Chemogenetic activation of MeAKiss neurons significantly increased LH in the absence of urine exposure, whereas inhibition of MeAKiss neurons did not alter LH. In situ hybridization revealed that MeAKiss neurons are a mixed neuronal population in which 71% express Vgat mRNA, 29% express Vglut2 mRNA, and 6% express both. CONCLUSIONS: Our results uncover, for the first time, that MeAKiss neurons process sexually relevant olfactory signals to influence reproductive hormone levels in male mice, likely through a complex interplay of neuropeptide and neurotransmitter signalling.


Assuntos
Tonsila do Cerebelo/fisiologia , Kisspeptinas/fisiologia , Hormônio Luteinizante/sangue , Neurônios/fisiologia , Feromônios/farmacologia , Administração por Inalação , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Clozapina/análogos & derivados , Clozapina/farmacologia , Ácido Glutâmico/metabolismo , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Transgênicos , Feromônios/administração & dosagem , Feromônios/urina , Área Pré-Óptica/fisiologia , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo
2.
Hum Mol Genet ; 25(9): 1771-9, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26908617

RESUMO

We recently reported the association of the PCSK6 gene with handedness through a quantitative genome-wide association study (GWAS; P < 0.5 × 10(-8)) for a relative hand skill measure in individuals with dyslexia. PCSK6 activates Nodal, a morphogen involved in regulating left-right body axis determination. Therefore, the GWAS data suggest that the biology underlying the patterning of structural asymmetries may also contribute to behavioural laterality, e.g. handedness. The association is further supported by an independent study reporting a variable number tandem repeat (VNTR) within the same PCSK6 locus to be associated with degree of handedness in a general population cohort. Here, we have conducted a functional analysis of the PCSK6 locus combining further genetic analysis, in silico predictions and molecular assays. We have shown that the previous GWAS signal was not tagging a VNTR effect, suggesting that the two markers have independent effects. We demonstrated experimentally that one of the top GWAS-associated markers, rs11855145, directly alters the binding site for a nuclear factor. Furthermore, we have shown that the predicted regulatory region adjacent to rs11855415 acts as a bidirectional promoter controlling the expression of novel RNA transcripts. These include both an antisense long non-coding RNA (lncRNA) and a short PCSK6 isoform predicted to be coding. This is the first molecular characterization of a handedness-associated locus that supports the role of common variants in non-coding sequences in influencing complex phenotypes through gene expression regulation.


Assuntos
Lateralidade Funcional/genética , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Íntrons/genética , Repetições Minissatélites/genética , Regiões Promotoras Genéticas/genética , Pró-Proteína Convertases/genética , Serina Endopeptidases/genética , Variação Genética/genética , Humanos , Proteína Nodal/genética , RNA Longo não Codificante/genética
3.
Mol Biol Evol ; 31(4): 765-78, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24361996

RESUMO

Gonadotropin-releasing hormone (GnRH) is a critical reproductive regulator in vertebrates. Homologous peptides are also found in invertebrates, with a variety of characterized functions. In the amphioxus, an invertebrate that provides the best model for the transition to vertebrates, four GnRH receptors (GnRHRs) were previously described, but their native ligands were not identified. Using a more sensitive search methodology with hidden Markov models, we identified the first GnRH-like peptide confirmed in the amphioxus Branchiostoma floridae. This peptide specifically activated one of the four GnRHRs. Although the primary structure of this peptide was divergent from any previously isolated GnRH peptide, the minimal conserved residues found in all other GnRH superfamily members were retained. The peptide was immunolocalized in proximity of the central canal of the anterior nerve cord, a region where other neuropeptides and receptors have been found. Additionally, the amphioxus GnRH-like gene was positioned in a locus surrounded by syntenic homologs of the human GnRH paralogon. The amphioxus GnRH-like peptide, with its distinct primary structure, activated a receptor with equal potency to multiple ligands that span the GnRH superfamily.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Anfioxos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Chlorocebus aethiops , Sequência Conservada , Evolução Molecular , Hormônio Liberador de Gonadotropina/química , Humanos , Dados de Sequência Molecular , Sistema Nervoso/metabolismo , Especificidade de Órgãos , Filogenia , Receptores LHRH/metabolismo , Transdução de Sinais , Sintenia
4.
N Engl J Med ; 366(7): 629-35, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-22335740

RESUMO

Gonadotropin-releasing hormone (GnRH) is the central regulator of gonadotropins, which stimulate gonadal function. Hypothalamic neurons that produce kisspeptin and neurokinin B stimulate GnRH release. Inactivating mutations in the genes encoding the human kisspeptin receptor (KISS1R, formerly called GPR54), neurokinin B (TAC3), and the neurokinin B receptor (TACR3) result in pubertal failure. However, human kisspeptin loss-of-function mutations have not been described, and contradictory findings have been reported in Kiss1-knockout mice. We describe an inactivating mutation in KISS1 in a large consanguineous family that results in failure of pubertal progression, indicating that functional kisspeptin is important for puberty and reproduction in humans. (Funded by the Scientific and Technological Research Council of Turkey [TÜBITAK] and others.).


Assuntos
Hipogonadismo/genética , Kisspeptinas/genética , Mutação , Puberdade/genética , Adolescente , Adulto , Criança , Consanguinidade , Feminino , Genes Recessivos , Técnicas de Genotipagem , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Masculino , Linhagem , Análise de Sequência de DNA
5.
Am J Physiol Endocrinol Metab ; 305(1): E67-77, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23632635

RESUMO

The human GnRH receptor (GNRHR1) has a specific set of properties with physiological and pharmacological influences not appropriately modeled in laboratory animals or cell-based systems. To address this deficiency, we have generated human GNRHR1 knock-in mice and described their reproductive phenotype. Measurement of pituitary GNRHR1 transcripts from homozygous human GNRHR1 knock-in (ki/ki) mice revealed a severe reduction (7- to 8-fold) compared with the mouse Gnrhr1 in wild-type mice. ¹²5I-GnRH binding assays on pituitary membrane fractions corroborated reduced human GNRHR1 protein expression in ki/ki mice, as occurs with transfection of human GNRHR1 in cell lines. Female homozygous knock-in mice displayed normal pubertal onset, indicating that a large reduction in GNRHR1 expression is sufficient for this process. However, ki/ki females exhibited periods of prolonged estrous and/or metestrous and reduced fertility. No impairment was found in reproductive maturity or adult fertility in male ki/ki mice. Interestingly, the serum LH response to GnRH challenge was reduced in both knock-in males and females, indicating a reduced GNRHR1 signaling capacity. Small molecules targeting human GPCRs usually have poor activities at homologous rodent receptors, thus limiting their use in preclinical development. Therefore, we tested a human-specific GnRH1 antagonist, NBI-42902, in our mouse model and demonstrated abrogation of a GnRH1-induced serum LH rise in ki/ki mice and an absence of effect in littermates expressing the wild-type murine receptor. This novel model provides the opportunity to study the human receptor in vivo and for screening the activity of human-specific GnRH analogs.


Assuntos
Ciclo Estral/fisiologia , Fertilidade/fisiologia , Receptores LHRH/genética , Receptores LHRH/fisiologia , Reprodução/fisiologia , Animais , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Feminino , Técnicas de Introdução de Genes , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Fenótipo , Hipófise/fisiologia , Gravidez , Receptores LHRH/antagonistas & inibidores , Maturidade Sexual/fisiologia , Testículo/crescimento & desenvolvimento , Testículo/fisiologia , Timina/análogos & derivados , Timina/farmacologia
6.
Neuroendocrinology ; 97(2): 193-202, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22377698

RESUMO

Pulsatile gonadotropin-releasing hormone (GnRH) is crucial to normal reproductive function and abnormalities in pulse frequency give rise to reproductive dysfunction. Kisspeptin and neurokinin B (NKB), neuropeptides secreted by the same neuronal population in the ventral hypothalamus, have emerged recently as critical central regulators of GnRH and thus gonadotropin secretion. Patients with mutations resulting in loss of signaling by either of these neuroendocrine peptides fail to advance through puberty but the mechanisms mediating this remain unresolved. We report here that continuous kisspeptin infusion restores gonadotropin pulsatility in patients with loss-of-function mutations in NKB (TAC3) or its receptor (TAC3R), indicating that kisspeptin on its own is sufficient to stimulate pulsatile GnRH secretion. Moreover, our findings suggest that NKB action is proximal to kisspeptin in the reproductive neuroendocrine cascade regulating GnRH secretion, and may act as an autocrine modulator of kisspeptin secretion. The ability of continuous kisspeptin infusion to induce pulsatile gonadotropin secretion further indicates that GnRH neurons are able to set up pulsatile secretion in the absence of pulsatile exogenous kisspeptin.


Assuntos
Transtornos do Desenvolvimento Sexual/genética , Kisspeptinas/administração & dosagem , Hormônio Luteinizante/metabolismo , Neurocinina B/deficiência , Receptores da Neurocinina-3/genética , Adulto , Transtornos do Desenvolvimento Sexual/sangue , Transtornos do Desenvolvimento Sexual/fisiopatologia , Transtornos do Desenvolvimento Sexual/terapia , Estradiol/sangue , Feminino , Humanos , Inibinas/sangue , Hormônio Luteinizante/sangue , Masculino , Mutação/fisiologia , Neurocinina B/genética , Fluxo Pulsátil/efeitos dos fármacos , Receptores da Neurocinina-3/deficiência , Receptores da Neurocinina-3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Testosterona/sangue
7.
Gen Comp Endocrinol ; 190: 134-43, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23756151

RESUMO

Diverse external and internal environmental factors are integrated in the hypothalamus to regulate the reproductive system. This is mediated through the pulsatile secretion of GnRH into the portal system to stimulate pituitary gonadotrophin secretion, which in turn regulates gonadal function. A single subpopulation of neurones termed 'KNDy neurones' located in the hypothalamic arcuate nucleus co-localise kisspeptin (Kiss), neurokinin B (NKB) and dynorphin (Dyn) and are responsive to negative feedback effects of sex steroids. The co-ordinated secretion from KNDy neurones appears to modulate the pulsatile release of GnRH, acting as a proximate pacemaker. This review briefly describes the neuropeptidergic control of reproduction in the avian class, highlighting the status of reproductive neuropeptide signalling systems homologous to those found in mammalian genomes. Genes encoding the GnRH system are complete in the chicken with similar roles to the mammalian counterparts, whereas genes encoding Kiss signalling components appear missing in the avian lineage, indicating a differing set of hypothalamic signals controlling avian reproduction. Gene sequences encoding both NKB and Dyn signalling components are present in the chicken genome, but expression analysis and functional studies remain to be completed. The focus of this article is to describe the avian complement of neuropeptidergic reproductive hormones and provide insights into the putative mechanisms that regulate reproduction in birds. These postulations highlight differences in reproductive strategies of birds in terms of gonadal steroid feedback systems, integration of metabolic signals and seasonality. Also included are propositions of KNDy neuropeptide gene silencing and plasticity in utilisation of these neuropeptides during avian evolution.


Assuntos
Galinhas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neuropeptídeos/metabolismo , Reprodução/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Dinorfinas/metabolismo , Kisspeptinas/metabolismo , Neurocinina B/metabolismo
8.
Ther Adv Endocrinol Metab ; 14: 20420188231173325, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37435528

RESUMO

Background: Endometriosis is an oestrogen-dependent disease that can cause subfertility in women who may require assisted reproductive technology (ART) to achieve their pregnancy goals. Objectives: The aim of this study was to compare ART outcomes in women with endometriosis following the long GnRH-agonist controlled ovarian stimulation (COS) protocol with those taking the GnRH-antagonist COS protocol. Data Sources and Methods: MEDLINE, Embase and Web of Science were systematically searched in June 2022. Randomized controlled trials (RCTs) and observational studies comparing the long GnRH-agonist COS protocol and the GnRH-antagonist COS protocol in women with all stages/subtypes of endometriosis were included. Data were synthesized into comprehensive tables for systematic review. The Scottish Intercollegiate Guidelines Network (SIGN) checklists were used for the risk of bias assessment of non-randomized studies and randomized studies, and all the included studies were deemed to have acceptable quality. Main Results: Eight studies (one RCT and seven observational) with 2695 patients (2761 cycles) were included. Most studies generally reported non-significant differences in clinical pregnancy or live birth rates regardless of the COS protocol used. However, the GnRH-agonist protocol may yield a higher total number of oocytes retrieved, especially mature oocytes. Conversely, the GnRH-antagonist protocol required a shorter COS duration and lower gonadotrophin dose. Adverse outcomes, such as rates of cycle cancellation and miscarriage, were similar between both COS protocols. Conclusion: Both the long GnRH-agonist and GnRH-antagonist COS protocols generally yield similar pregnancy outcomes. However, the long GnRH-agonist protocol may be associated with a higher cumulative pregnancy rate due to the higher number of retrieved oocytes available for cryopreservation. The underlying mechanisms of the two COS protocols on the female reproductive tract remain unclear. Clinicians should consider treatment costs, stage/subtype of endometriosis and pregnancy goals of their patients when selecting a GnRH analogue for COS. A well-powered RCT is needed to minimize the risk of bias and compare the risk for ovarian hyperstimulation syndrome. Registration: This review was prospectively registered at PROSPERO under Registration No. CRD42022327604.

9.
N Engl J Med ; 360(26): 2742-8, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19535795

RESUMO

We investigated whether mutations in the gene encoding gonadotropin-releasing hormone 1 (GNRH1) might be responsible for idiopathic hypogonadotropic hypogonadism (IHH) in humans. We identified a homozygous GNRH1 frameshift mutation, an insertion of an adenine at nucleotide position 18 (c.18-19insA), in the sequence encoding the N-terminal region of the signal peptide-containing protein precursor of gonadotropin-releasing hormone (prepro-GnRH) in a teenage brother and sister, who had normosmic IHH. Their unaffected parents and a sibling who was tested were heterozygous. This mutation results in an aberrant peptide lacking the conserved GnRH decapeptide sequence, as shown by the absence of immunoreactive GnRH when expressed in vitro. This isolated autosomal recessive GnRH deficiency, reversed by pulsatile GnRH administration, shows the pivotal role of GnRH in human reproduction.


Assuntos
Mutação da Fase de Leitura , Hormônio Liberador de Gonadotropina/genética , Hipogonadismo/genética , Precursores de Proteínas/genética , Adolescente , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Genes Recessivos , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/deficiência , Homozigoto , Humanos , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Masculino , Linhagem , Análise de Sequência de DNA , Testosterona/sangue
10.
Adv Ther ; 38(10): 5025-5045, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34514552

RESUMO

INTRODUCTION: Hot flushes/flashes (HFs) or other vasomotor symptoms affect between 45 and 97% of women during menopause. Hormone replacement therapy (HRT) is effective at alleviating menopausal symptoms, but some women cannot or prefer not to take HRT. Since current non-hormonal options have suboptimal efficacy/tolerability, there is a pressing need for an effective, well-tolerated alternative. The neurokinin 3 receptor (NK3R) has recently been implicated in the generation of menopausal HFs and represents a novel therapeutic target to ameliorate HF symptoms. This review aims to assess if NK3R antagonists (NK3Ras) are more effective than Serotonin Norepinephrine Reuptake Inhibitors (SNRIs)-currently a common choice for non-hormonal treatment of menopausal HFs. METHODS: Studies were identified after systematically searching Ovid MEDLINE and EMBASE databases based on PRISMA guidelines. Trial quality and bias were assessed. Key efficacy outcomes (HF frequency, HF severity and number of night-time awakenings/night-sweats) and selected safety outcomes were extracted and analysed. RESULTS: Seven SNRI and four NK3Ra placebo-controlled randomised trials (plus four follow-up reports) were included in this review. NK3Ra administration resulted in a larger reduction from baseline in HF frequency, HF severity and night-sweats compared to SNRIs. Five of seven SNRI trials showed a reduction in HF frequency that was statistically significant (by 48-67% from baseline at weeks 8 or 12) whereas all NK3Ra trials showed a statistically significant reduction in HF frequency (by 62-93% from baseline at weeks 2, 4 or 12). While SNRI trials reported poor tolerability, particularly nausea, NK3Ra trials reported good tolerability overall, although two trials reported elevation in transaminases. CONCLUSION: NK3Ras trials show encouraging efficacy and tolerability/safety. Completion of phase 3 NK3Ra trials are required to confirm efficacy and uphold safety/tolerability data but phase 2 results suggest that NK3Ras are more effective than SNRIs for non-hormonal treatment of menopausal HFs.


Assuntos
Inibidores Seletivos de Recaptação de Serotonina , Serotonina , Feminino , Humanos , Menopausa , Norepinefrina , Receptores da Neurocinina-3 , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico
11.
J Neuroendocrinol ; 32(2): e12819, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31800973

RESUMO

The present study investigated neuroanatomically localised changes in de novo DNA methyltransferase expression in the female Siberian hamster (Phodopus sungorus). The objectives were to identify the neuroendocrine substrates that exhibit rhythmic Dnmt3a and Dnmt3b expression across the oestrous cycle and also examine the role of ovarian steroids. Hypothalamic Dnmt3a expression was observed to significantly increase during the transition from pro-oestrous to oestrous. A single bolus injection of diethylstilbestrol and progesterone was sufficient to increase Dnmt3a cell numbers and Dnmt3b immunoreactive intensity in the suprachiasmatic nucleus. In vitro analyses using an embryonic rodent cell line revealed that diethylstilbestrol was sufficient to induce Dnmt3b expression. Up-regulating DNA methylation in vitro reduced the expression of vasoactive intestinal polypeptide, Vip, and the circadian clock gene, Bmal1. Together, these data indicate that ovarian steroids drive de novo DNA methyltransferase expression in the mammalian suprachiasmatic nucleus and increased methylation may regulate genes involved in the circadian timing of oestrous: Vip and Bmal1. Overall, epigenetically mediated neuroendocrine reproductive events may reflect an evolutionarily ancient process involved in the timing of female fertility.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Hormônios Gonadais/metabolismo , Ovário/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Relógios Circadianos , Metilação de DNA , Ciclo Estral/metabolismo , Feminino , Sistemas Neurossecretores/metabolismo , Phodopus
12.
Adv Biosyst ; 3(3): e1800290, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-32627397

RESUMO

Optogenetics, photostimulation of neural tissues rendered sensitive to light, is widely used in neuroscience to modulate the electrical excitability of neurons. For effective optical excitation of neurons, light wavelength and power density must fit with the expression levels and biophysical properties of the genetically encoded light-sensitive ion channels used to confer light sensitivity on cells-most commonly, channelrhodopsins (ChRs). As light sources, organic light-emitting diodes (OLEDs) offer attractive properties for miniaturized implantable devices for in vivo optical stimulation, but they do not yet operate routinely at the optical powers required for optogenetics. Here, OLEDs with doped charge transport layers are demonstrated that deliver blue light with good stability over millions of pulses, at powers sufficient to activate the ChR, CheRiff when expressed in cultured primary neurons, allowing live cell imaging of neural activity with the red genetically encoded calcium indicator, jRCaMP1a. Intracellular calcium responses scale with the radiant flux of OLED emission, when varied through changes in the current density, number of pulses, frequency, and pulse width delivered to the devices. The reported optimization and characterization of high-power OLEDs are foundational for the development of miniaturized OLEDs with thin-layer encapsulation on bioimplantable devices to allow single-cell activation in vivo.


Assuntos
Neurônios , Optogenética/métodos , Estimulação Luminosa/métodos , Animais , Células Cultivadas , Channelrhodopsins/química , Channelrhodopsins/metabolismo , Eletrodos Implantados , Hipocampo/citologia , Camundongos , Neurônios/química , Neurônios/citologia , Neurônios/metabolismo , Proteínas Recombinantes
13.
J Comp Neurol ; 527(16): 2634-2643, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30950042

RESUMO

Dyslexia is a common neurodevelopmental disorder caused by a significant genetic component. The KIAA0319 gene is one of the most robust dyslexia susceptibility factors but its function remains poorly understood. Initial RNA-interference studies in rats suggested a role in neuronal migration whereas subsequent work with double knock-out mouse models for both Kiaa0319 and its paralogue Kiaa0319-like reported effects in the auditory system but not in neuronal migration. To further understand the role of KIAA0319 during neurodevelopment, we carried out an expression study of its zebrafish orthologue at different embryonic stages. We used different approaches including RNAscope in situ hybridization combined with light-sheet microscopy. The results show particularly high expression during the first few hours of development. Later, expression becomes localized in well-defined structures. In addition to high expression in the brain, we report for the first time expression in the eyes and the notochord. Surprisingly, kiaa0319-like, which generally shows a similar expression pattern to kiaa0319, was not expressed in the notochord suggesting a distinct role for kiaa0319 in this structure. This observation was supported by the identification of notochord enhancers enriched upstream of the KIAA0319 transcription start site, in both zebrafish and humans. This study supports a developmental role for KIAA0319 in the brain as well as in other developing structures, particularly in the notochord which, is key for establishing body patterning in vertebrates.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Olho/embriologia , Olho/metabolismo , Notocorda/metabolismo , Animais , Animais Geneticamente Modificados , Movimento Celular/fisiologia , Dislexia/genética , Dislexia/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fígado/metabolismo , Miocárdio/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Peixe-Zebra
14.
Sci Adv ; 4(4): eaar4817, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29740614

RESUMO

Scattering and absorption limit the penetration of optical fields into tissue. We demonstrate a new approach for increased depth penetration in light-sheet microscopy: attenuation-compensation of the light field. This tailors an exponential intensity increase along the illuminating propagation-invariant field, enabling the redistribution of intensity strategically within a sample to maximize signal and minimize irradiation. A key attribute of this method is that only minimal knowledge of the specimen transmission properties is required. We numerically quantify the imaging capabilities of attenuation-compensated Airy and Bessel light sheets, showing that increased depth penetration is gained without compromising any other beam attributes. This powerful yet straightforward concept, combined with the self-healing properties of the propagation-invariant field, improves the contrast-to-noise ratio of light-sheet microscopy up to eightfold across the entire field of view in thick biological specimens. This improvement can significantly increase the imaging capabilities of light-sheet microscopy techniques using Airy, Bessel, and other propagation-invariant beam types, paving the way for widespread uptake by the biomedical community.

15.
Biomed Opt Express ; 8(11): 5179-5190, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29188112

RESUMO

Optical clearing is emerging as a popular approach particularly for studies in neuroscience. However the use of corrosive clearing solutions typically requires sophisticated objectives or extreme care with optical components chosen for single- or multi-photon imaging. In contrast to the use of complex, custom-made microscope objectives, we show that the use of a corrected multimode fibre (MMF) offers a route that is resistant to corrosion, can be used in clearing media, is not constrained by the refractive index of the immersion medium and offers flexible working distances. Using a corrected MMF, we demonstrate fluorescence imaging of beads and stained neuroblastoma cells through optically cleared mouse brain tissue, as well as imaging in an extreme oxidative environment to show the versatility of our approach. Additionally, we perform Raman imaging of polystyrene beads in clearing media to demonstrate that this approach may be used for vibrational spectroscopy of optically cleared samples.

16.
Biomed Opt Express ; 7(10): 4021-4033, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27867712

RESUMO

We have investigated the effect of Airy illumination on the image quality and depth penetration of digitally scanned light-sheet microscopy in turbid neural tissue. We used Fourier analysis of images acquired using Gaussian and Airy light-sheets to assess their respective image quality versus penetration into the tissue. We observed a three-fold average improvement in image quality at 50 µm depth with the Airy light-sheet. We also used optical clearing to tune the scattering properties of the tissue and found that the improvement when using an Airy light-sheet is greater in the presence of stronger sample-induced aberrations. Finally, we used homogeneous resolution probes in these tissues to quantify absolute depth penetration in cleared samples with each beam type. The Airy light-sheet method extended depth penetration by 30% compared to a Gaussian light-sheet.

17.
Endocrinology ; 146(9): 4061-73, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15961566

RESUMO

In vertebrates, GnRH binds to its receptor and stimulates predominantly G(q/11)-mediated signal transduction in gonadotropes. However, little is known about the GnRH receptor and its signaling pathway in tunicates, a group that arose before the vertebrates. Although tunicates have had duplications of a few genes in the last 600 million years, the early vertebrates had duplications of the full genome. Also unknown is the nature of GnRH signaling in the tunicate, which lacks both a pituitary gland and sex steroids. However, we know that tunicates have GnRH peptides because we previously reported six GnRH peptides encoded within the tunicate genome of Ciona intestinalis. Here we clone and sequence cDNAs for four putative GnRH receptors from C. intestinalis. These are the only invertebrate GnRH receptors found to date. Each Ciona GnRH receptor was expressed in COS-7 cells, incubated with each of the six C. intestinalis GnRHs and assayed for a signaling response. GnRH receptors 1, 2, and 3 responded to Ciona GnRH peptides to stimulate intracellular cAMP accumulation. In contrast, only GnRH receptor 1 activated inositol phosphate turnover in response to one of the Ciona GnRHs. The green monkey type II GnRH receptor cDNA was tested as a comparison and a positive control. In conclusion, the four GnRH receptors encoded within the C. intestinalis genome were all transcribed into messenger RNA, but only three of the Ciona GnRH receptors were biologically active in our assays. The Ciona GnRH receptors almost exclusively activated the cAMP pathway.


Assuntos
Ciona intestinalis/genética , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Sequência Consenso , AMP Cíclico/metabolismo , Éxons , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , Receptores LHRH/química , Transdução de Sinais/fisiologia
18.
Mol Cell Endocrinol ; 231(1-2): 49-63, 2005 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-15713535

RESUMO

To understand the role of growth hormone-releasing hormone (GHRH) and pituitary adenylate cyclase-activating polypeptide (PACAP) and to examine the functional significance of the co-expression of GHRH and PACAP in fish, their receptors were characterized in zebrafish. Three cDNAs encoding the PAC(1) receptor, the VPAC(1) receptor, and the partial GHRH receptor were identified from zebrafish. Functional expression of the PAC(1) and VPAC(1) receptors revealed that both are potently coupled to the adenylyl cyclase pathway, but only the PAC(1) receptor is coupled to the phospholipase C pathway. Transcripts for all three receptors were widely distributed, often in an overlapping pattern in the adult zebrafish. Also, one splice variant of the partial GHRH receptor and three splice variants of the PAC(1) receptor were identified from adult zebrafish. The long GHRH receptor transcript contained a 27 amino acid insert in transmembrane domain 5 encoding a premature stop codon leading to a truncated receptor protein. For the PAC(1) receptor, two of the splice variants corresponded to the hop1 and hop2 variants characterized in mammals. The third splice variant identified from the gill encoded a novel 107 bp insert containing a premature stop codon. Therefore, PACAP and GHRH have widespread, overlapping target sites suggesting a coordinated role for these hormones in evolution.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/genética , Receptores de Superfície Celular/genética , Receptores de Peptídeo Intestinal Vasoativo/genética , Adenilil Ciclases/metabolismo , Animais , Sequência de Bases , Códon sem Sentido , DNA Complementar , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Dados de Sequência Molecular , Splicing de RNA , RNA Mensageiro/análise , Receptores de Superfície Celular/fisiologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores de Peptídeo Intestinal Vasoativo/fisiologia , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo , Alinhamento de Sequência , Distribuição Tecidual , Fosfolipases Tipo C/metabolismo , Peixe-Zebra
19.
Endocrinology ; 144(5): 1907-19, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12697698

RESUMO

GnRH is the key regulator of the reproductive axis in vertebrates, but little is known about GnRH before the origin of vertebrates. We have identified two genes encoding GnRH in a protochordate, Ciona intestinalis, thought to be related to the ancestral animal that gave rise to vertebrates. Each gene, Ci-gnrh1 and Ci-gnrh2, encodes in tandem three GnRH peptides, each of which is unique compared with known forms. Ci-gnrh1 encodes three peptides and contains no introns, whereas Ci-gnrh2 encodes three more peptides but has two introns. This is the first report in which more than one GnRH peptide is encoded on a single gene. The Ciona genes reveal consensus promoter elements that are conserved compared with human GNRH1. Both tunicate genes are expressed as mRNA early and throughout development, measured at the stages of four-cell, gastrulation, tail release, and tail resorption. In a closely related tunicate species, Ciona savignyi, we used in silico analysis to identify two similar genes encoding six peptides, only one of which is unique compared with C. intestinalis. Immunohistochemistry showed that at least one GnRH peptide was in the nerve net that surrounds the dorsal strand. Synthetic forms of the seven novel tunicate peptides induced release of gametes in adult tunicates. In contrast, the peptides did not activate the human GnRH-I receptor or cause release of LH in a rat pituitary cell assay. These data provide insight into the structural evolution of the GnRH peptides and their genes and show a functional role for GnRH in tunicate spawning.


Assuntos
Ciona intestinalis/genética , Hormônio Liberador de Gonadotropina/genética , Envelhecimento/fisiologia , Sequência de Aminoácidos/genética , Animais , Sítios de Ligação/genética , Ciona intestinalis/crescimento & desenvolvimento , Ciona intestinalis/metabolismo , Reações Cruzadas , Células Germinativas/fisiologia , Hormônio Liberador de Gonadotropina/imunologia , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/metabolismo , Ratos , Receptores LHRH/metabolismo , Fatores de Transcrição/genética , Sítio de Iniciação de Transcrição , Repetições de Trinucleotídeos
20.
PLoS One ; 7(6): e38456, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22679506

RESUMO

Congenital hypogonadotropic hypogonadism (CHH) is characterized by low gonadotropins and failure to progress normally through puberty. Mutations in the gene encoding the GnRH receptor (GNRHR1) result in CHH when present as compound heterozygous or homozygous inactivating mutations. This study identifies and characterizes the properties of two novel GNRHR1 mutations in a family in which three brothers display normosmic CHH while their sister was unaffected. Molecular analysis in the proband and the affected brothers revealed two novel non-synonymous missense GNRHR1 mutations, present in a compound heterozygous state, whereas their unaffected parents possessed only one inactivating mutation, demonstrating the autosomal recessive transmission in this kindred and excluding X-linked inheritance equivocally suggested by the initial pedigree analysis. The first mutation at c.845 C>G introduces an Arg substitution for the conserved Pro 282 in transmembrane domain (TMD) 6. The Pro282Arg mutant is unable to bind radiolabeled GnRH analogue. As this conserved residue is important in receptor conformation, it is likely that the mutation perturbs the binding pocket and affects trafficking to the cell surface. The second mutation at c.968 A>G introduces a Cys substitution for Tyr 323 in the functionally crucial N/DPxxY motif in TMD 7. The Tyr323Cys mutant has an increased GnRH binding affinity but reduced receptor expression at the plasma membrane and impaired G protein-coupling. Inositol phosphate accumulation assays demonstrated absent and impaired Gα(q/11) signal transduction by Pro282Arg and Tyr323Cys mutants, respectively. Pretreatment with the membrane permeant GnRHR antagonist NBI-42902, which rescues cell surface expression of many GNRHR1 mutants, significantly increased the levels of radioligand binding and intracellular signaling of the Tyr323Cys mutant but not Pro282Arg. Immunocytochemistry confirmed that both mutants are present on the cell membrane albeit at low levels. Together these molecular deficiencies of the two novel GNRHR1 mutations lead to the CHH phenotype when present as a compound heterozygote.


Assuntos
Hipogonadismo/genética , Receptores LHRH/genética , Adolescente , Adulto , Animais , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Feminino , Heterozigoto , Humanos , Hipogonadismo/congênito , Hipogonadismo/metabolismo , Fosfatos de Inositol/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA