Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Reprod ; 36(7): 1891-1906, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34059912

RESUMEN

STUDY QUESTION: Does hypo-glycosylated human recombinant FSH (hFSH18/21) have greater in vivo bioactivity that drives follicle development in vivo compared to fully-glycosylated human recombinant FSH (hFSH24)? SUMMARY ANSWER: Compared with fully-glycosylated hFSH, hypo-glycosylated hFSH has greater bioactivity, enabling greater follicular health and growth in vivo, with enhanced transcriptional activity, greater activation of receptor tyrosine kinases (RTKs) and elevated phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and Mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling. WHAT IS KNOWN ALREADY: Glycosylation of FSH is necessary for FSH to effectively activate the FSH receptor (FSHR) and promote preantral follicular growth and formation of antral follicles. In vitro studies demonstrate that compared to fully-glycosylated recombinant human FSH, hypo-glycosylated FSH has greater activity in receptor binding studies, and more effectively stimulates the PKA pathway and steroidogenesis in human granulosa cells. STUDY DESIGN, SIZE, DURATION: This is a cross-sectional study evaluating the actions of purified recombinant human FSH glycoforms on parameters of follicular development, gene expression and cell signaling in immature postnatal day (PND) 17 female CD-1 mice. To stimulate follicle development in vivo, PND 17 female CD-1 mice (n = 8-10/group) were treated with PBS (150 µl), hFSH18/21 (1 µg/150 µl PBS) or hFSH24 (1 µg/150 µl PBS) by intraperitoneal injection (i.p.) twice daily (8:00 a.m. and 6:00 p.m.) for 2 days. Follicle numbers, serum anti-Müllerian hormone (AMH) and estradiol levels, and follicle health were quantified. PND 17 female CD-1 mice were also treated acutely (2 h) in vivo with PBS, hFSH18/21 (1 µg) or hFSH24 (1 µg) (n = 3-4/group). One ovary from each mouse was processed for RNA sequencing analysis and the other ovary processed for signal transduction analysis. An in vitro ovary culture system was used to confirm the relative signaling pathways. PARTICIPANTS/MATERIALS, SETTING, METHODS: The purity of different recombinant hFSH glycoforms was analyzed using an automated western blot system. Follicle numbers were determined by counting serial sections of the mouse ovary. Real-time quantitative RT-PCR, western blot and immunofluorescence staining were used to determine growth and apoptosis markers related with follicle health. RNA sequencing and bioinformatics were used to identify pathways and processes associated with gene expression profiles induced by acute FSH glycoform treatment. Analysis of RTKs was used to determine potential FSH downstream signaling pathways in vivo. Western blot and in vitro ovarian culture system were used to validate the relative signaling pathways. MAIN RESULTS AND THE ROLE OF CHANCE: Our present study shows that both hypo- and fully-glycosylated recombinant human FSH can drive follicular growth in vivo. However, hFSH18/21 promoted development of significantly more large antral follicles compared to hFSH24 (P < 0.01). In addition, compared with hFSH24, hFSH18/21 also promoted greater indices of follicular health, as defined by lower BAX/BCL2 ratios and reduced cleaved Caspase 3. Following acute in vivo treatment with FSH glycoforms RNA-sequencing data revealed that both FSH glycoforms rapidly induced ovarian transcription in vivo, but hypo-glycosylated FSH more robustly stimulated Gαs and cAMP-mediated signaling and members of the AP-1 transcription factor complex. Moreover, hFSH18/21 treatment induced significantly greater activation of RTKs, PI3K/AKT and MAPK/ERK signaling compared to hFSH24. FSH-induced indices of follicle growth in vitro were blocked by inhibition of PI3K and MAPK. LARGE SCALE DATA: RNA sequencing of mouse ovaries. Data will be shared upon reasonable request to the corresponding author. LIMITATIONS, REASONS FOR CAUTION: The observations that hFSH glycoforms have different bioactivities in the present study employing a mouse model of follicle development should be verified in nonhuman primates. The gene expression studies reflect transcriptomes of whole ovaries. WIDER IMPLICATIONS OF THE FINDINGS: Commercially prepared recombinant human FSH used for ovarian stimulation in human ART is fully-glycosylated FSH. Our findings that hypo-glycosylated hFSH has greater bioactivity enabling greater follicular health and growth without exaggerated estradiol production in vivo, demonstrate the potential for its development for application in human ART. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by NIH 1P01 AG029531, NIH 1R01 HD 092263, VA I01 BX004272, and the Olson Center for Women's Health. JSD is the recipient of a VA Senior Research Career Scientist Award (1IK6 BX005797). This work was also partially supported by National Natural Science Foundation of China (No. 31872352). The authors declared there are no conflicts of interest.


Asunto(s)
Hormona Folículo Estimulante Humana , Proteínas Quinasas Activadas por Mitógenos , Folículo Ovárico/crecimiento & desarrollo , Fosfatidilinositol 3-Quinasas , Transducción de Señal , Animales , China , Estudios Transversales , Femenino , Glicosilación , Ratones , Proteínas Recombinantes
2.
Mol Hum Reprod ; 26(12): 894-905, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33084890

RESUMEN

Commercial hMG drugs are marketed for the treatment of infertility and consist of highly purified hormones acting on receptors expressed in target gonadal cells. Menopur® and Meriofert® are combined preparation of FSH and hCG and are compared in vitro herein. To this purpose, the molecular composition of the two drugs was analyzed by immunoassay. The formation of FSH receptor and LH/hCG receptor (FSHR; LHCGR) heteromer, intracellular Ca2+ and cAMP activation, ß-arrestin 2 recruitment and the synthesis of progesterone and estradiol were evaluated in transfected HEK293 and human primary granulosa lutein cells treated by drugs administered within the pg-mg/ml concentration range. Molecular characterization revealed that Meriofert® has a higher FSH:hCG ratio than Menopur® which, in turn, displays the presence of LH molecules. While both drugs induced similar FSHR-LHCGR heteromeric formations and intracellular Ca2+ increase, Meriofert® had a higher potency than Menopur® in inducing a cAMP increase. Moreover, Meriofert® revealed a higher potency than Menopur® in recruiting ß-arrestin 2, likely due to different FSH content modulating the tridimensional structure of FSHR-LHCGR-ß-arrestin 2 complexes, as evidenced by a decrease in bioluminescence resonance energy transfer signal. This drug-specific activation of intracellular signaling pathways is consistent with the molecular composition of these preparations and impacts downstream progesterone and estradiol production, with Menopur® more potent than Meriofert® in inducing the synthesis of both the steroids. These findings are suggestive of distinct in-vivo activities of these preparations, but require cautious interpretation and further validation from clinical studies.


Asunto(s)
Gonadotropinas/metabolismo , Menopausia/fisiología , Calcio/metabolismo , Gonadotropina Coriónica/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Células HEK293 , Humanos , Inmunoensayo , Transducción de Señal/genética , Transducción de Señal/fisiología
3.
Cell Tissue Res ; 369(3): 567-578, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28451751

RESUMEN

The natriuretic peptides, Atrial-, B-type and C-type natriuretric peptides (ANP, BNP, CNP), are regulators of many endocrine tissues and exert their effects predominantly through the activation of their specific guanylyl cyclase receptors (GC-A and GC-B) to generate cGMP. Whereas cGMP-independent signalling has been reported in response to natriuretic peptides, this is mediated via either the clearance receptor (Npr-C) or a renal-specific NPR-Bi isoform, which both lack intrinsic guanylyl cyclase activity. Here, we report evidence of GC-B-dependent cGMP-independent signalling in pituitary GH3 cells. Stimulation of GH3 cells with CNP resulted in a rapid and sustained enhancement of ERK1/2 phosphorylation (P-ERK1/2), an effect that was not mimicked by dibutryl-cGMP. Furthermore, CNP-stimulated P-ERK1/2 occurred at concentrations below that required for cGMP accumulation. The effect of CNP on P-ERK1/2 was sensitive to pharmacological blockade of MEK (U0126) and Src kinases (PP2). Silencing of the GC-B1 and GC-B2 splice variants of the GC-B receptor by using targeted short interfering RNAs completely blocked the CNP effects on P-ERK1/2. CNP failed to alter GH3 cell proliferation or cell cycle distribution but caused a concentration-dependent increase in the activity of the human glycoprotein α-subunit promoter (αGSU) in a MEK-dependent manner. Finally, CNP also activated the p38 and JNK MAPK pathways in GH3 cells. These findings reveal an additional mechanism of GC-B signalling and suggest additional biological roles for CNP in its target tissues.


Asunto(s)
Guanilato Ciclasa/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Péptido Natriurético Tipo-C/farmacología , Somatotrofos/metabolismo , Animales , Línea Celular , GMP Cíclico/metabolismo , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Somatotrofos/efectos de los fármacos
4.
J Biol Chem ; 290(7): 3875-92, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25516594

RESUMEN

Formation of G protein-coupled receptors (GPCRs) into dimers and higher order oligomers represents a key mechanism in pleiotropic signaling, yet how individual protomers function within oligomers remains poorly understood. We present a super-resolution imaging approach, resolving single GPCR molecules to ∼ 8 nm resolution in functional asymmetric dimers and oligomers using dual-color photoactivatable dyes and localization microscopy (PD-PALM). PD-PALM of two functionally defined mutant luteinizing hormone receptors (LHRs), a ligand-binding deficient receptor (LHR(B-)) and a signaling-deficient (LHR(S-)) receptor, which only function via intermolecular cooperation, favored oligomeric over dimeric formation. PD-PALM imaging of trimers and tetramers revealed specific spatial organizations of individual protomers in complexes where the ratiometric composition of LHR(B-) to LHR(S-) modulated ligand-induced signal sensitivity. Structural modeling of asymmetric LHR oligomers strongly aligned with PD-PALM-imaged spatial arrangements, identifying multiple possible helix interfaces mediating inter-protomer associations. Our findings reveal that diverse spatial and structural assemblies mediating GPCR oligomerization may acutely fine-tune the cellular signaling profile.


Asunto(s)
Colorantes Fluorescentes/química , Procesamiento de Imagen Asistido por Computador , Multimerización de Proteína , Receptores de HL/química , Receptores de HL/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Espectrometría de Fluorescencia , Espectrofotometría Ultravioleta
5.
Reproduction ; 148(4): R63-70, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25030890

RESUMEN

The advent of technologies to genetically manipulate the mouse genome has revolutionised research approaches, providing a unique platform to study the causality of reproductive disorders in vivo. With the relative ease of generating genetically modified (GM) mouse models, the last two decades have yielded multiple loss-of-function and gain-of-function mutation mouse models to explore the role of gonadotrophins and their receptors in reproductive pathologies. This work has provided key insights into the molecular mechanisms underlying reproductive disorders with altered gonadotrophin action, revealing the fundamental roles of these pituitary hormones and their receptors in the hypothalamic-pituitary-gonadal axis. This review will describe GM mouse models of gonadotrophins and their receptors with enhanced or diminished actions, specifically focusing on the male. We will discuss the mechanistic insights gained from these models into male reproductive disorders, and the relationship and understanding provided into male human reproductive disorders originating from altered gonadotrophin action.


Asunto(s)
Gonadotropinas/metabolismo , Infertilidad Masculina/metabolismo , Reproducción , Animales , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/metabolismo , Genotipo , Gonadotropinas/genética , Infertilidad Masculina/genética , Infertilidad Masculina/fisiopatología , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Reproducción/genética , Transducción de Señal
6.
Mol Hum Reprod ; 18(8): 379-90, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22554618

RESUMEN

Heterodimeric hCG is one of the key hormones determining early pregnancy success. We have previously identified rare missense mutations in hCGß genes with potential pathophysiological importance. The present study assessed the impact of these mutations on the structure and function of hCG by applying a combination of in silico (sequence and structure analysis, molecular dynamics) and in vitro (co-immunoprecipitation, immuno- and bioassays) approaches. The carrier status of each mutation was determined for 1086 North-Europeans [655 patients with recurrent miscarriage (RM)/431 healthy controls from Estonia, Finland and Denmark] using PCR-restriction fragment length polymorphism. The mutation CGB5 p.Val56Leu (rs72556325) was identified in a single heterozygous RM patient and caused a structural hindrance in the formation of the hCGα/ß dimer. Although the amount of the mutant hCGß assembled into secreted intact hCG was only 10% compared with the wild-type, a stronger signaling response was triggered upon binding to its receptor, thus compensating the effect of poor dimerization. The mutation CGB8 p.Pro73Arg (rs72556345) was found in five heterozygotes (three RM cases and two control individuals) and was inherited by two of seven studied live born children. The mutation caused ~50% of secreted ß-subunits to acquire an alternative conformation, but did not affect its biological activity. For the CGB8 p.Arg8Trp (rs72556341) substitution, the applied in vitro methods revealed no alterations in the assembly of intact hCG as also supported by an in silico analysis. In summary, the accumulated data indicate that only mutations with neutral or mild functional consequences might be tolerated in the major hCGß genes CGB5 and CGB8.


Asunto(s)
Aborto Habitual/genética , Gonadotropina Coriónica Humana de Subunidad beta/genética , Mutación Missense , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células CHO , Línea Celular , Gonadotropina Coriónica Humana de Subunidad beta/química , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Cricetinae , Femenino , Humanos , Simulación de Dinámica Molecular , Embarazo , Complicaciones del Embarazo/genética , Conformación Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Análisis de Secuencia de ADN
7.
Endocrinology ; 163(12)2022 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-36201606

RESUMEN

Follicle-stimulating hormone (FSH) is a key endocrine regulator of ovarian function. FSH is secreted as 2 macroglycosylation variants: partially glycosylated FSH (FSH21/18) and fully glycosylated FSH (FSH24). FSH21/18 is more potent than FSH24 at binding to and activating the FSH receptor (R). The ratio of FSH21/18:FSH24 has been shown to change with age, with FSH21/18 predominant at reproductive prime, and FSH24 predominant during perimenopause/menopause. How these FSH glycosylation variants modulate ovarian follicle functions remains largely unknown. The aim of this study was to investigate the effect of FSH glycosylation variants of pre-antral follicle function. Pre-antral follicles were isolated from 3- to 5-week-old C57BL/6 mice and treated ±10 ng/mL FSH21/18, FSH24, a ratio of 80:20 FSH21/18:FSH24 (to mimic reproductive prime), 50:50 FSH21/18:FSH24 (perimenopause), or 20:80 FSH21/18:FSH24 (menopause) for up to 96 hours. FSH21/18 and 80:20 FSH21/18:FSH24 increased follicle growth, in comparison with control, contrasting with FSH24 and 20:80 FSH21/18:FSH24. Survival rates were decreased in follicles treated with FSH24 or 20:80 FSH21/18:FSH24, with follicles undergoing basement membrane rupture and oocyte extrusion, increased Caspase3 gene and protein expression, and decreased markers of cell proliferation in FSH24 or 20:80 FSH21/18:FSH24-treated follicles. Moreover, this correlated with differential regulation of key genes modulating follicular functions. Pharmacological inhibitors of key FSH signal pathways suggests FSH21/18 and FSH24 initiate different FSHR signal pathway activation, which may determine their differential effects on follicle growth and survival. These data suggest that the nature of FSH glycosylation modulates the follicular cellular environment to regulate follicle growth and survival and may underpin the increasing ovarian resistance to FSH observed during aging.


Asunto(s)
Hormona Folículo Estimulante , Receptores de HFE , Femenino , Ratones , Animales , Hormona Folículo Estimulante/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Glicosilación , Ratones Endogámicos C57BL , Folículo Ovárico/metabolismo
8.
Front Endocrinol (Lausanne) ; 12: 765727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925235

RESUMEN

Follicle-stimulating hormone (FSH) and its target G protein-coupled receptor (FSHR) are essential for reproduction. Recent studies have established that the hypo-glycosylated pituitary FSH glycoform (FSH21/18), is more bioactive in vitro and in vivo than the fully-glycosylated variant (FSH24). FSH21/18 predominates in women of reproductive prime and FSH24 in peri-post-menopausal women, suggesting distinct functional roles of these FSH glycoforms. The aim of this study was to determine if differential FSH glycosylation modulated FSHR oligomerization and resulting impact on cAMP signaling. Using a modified super-resolution imaging technique (PD-PALM) to assess FSHR complexes in HEK293 cells expressing FSHR, we observed time and concentration-dependent modulation of FSHR oligomerization by FSH glycoforms. High eFSH and FSH21/18 concentrations rapidly dissociated FSHR oligomers into monomers, whereas FSH24 displayed slower kinetics. The FSHR ß-arrestin biased agonist, truncated eLHß (Δ121-149) combined with asparagine56-deglycosylated eLHα (dg-eLHt), increased FSHR homomerization. In contrast, low FSH21/18 and FSH24 concentrations promoted FSHR association into oligomers. Dissociation of FSHR oligomers correlated with time points where higher cAMP production was observed. Taken together, these data suggest that FSH glycosylation may modulate the kinetics and amplitude of cAMP production, in part, by forming distinct FSHR complexes, highlighting potential avenues for novel therapeutic targeting of the FSHR to improve IVF outcomes.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Receptores de HFE/metabolismo , Transducción de Señal/fisiología , Línea Celular , Glicosilación , Células HEK293 , Humanos
9.
Methods Mol Biol ; 1947: 337-348, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30969426

RESUMEN

Super-resolution imaging has provided unprecedented insight in the molecular complexities of fundamental cell biological questions. For G protein-coupled receptors (GPCRs), its application to the study of receptor homomers and heteromers have unveiled the diversity of complexes these GPCRs can form at the plasma membrane at a structural and functional level. Here, we describe our methodological approach of photoactivated localization microscopy with photoactivatable dyes (PD-PALM) to visualize and quantify the spatial assembly of GPCR heteromers at the plasma membrane.


Asunto(s)
Membrana Celular/metabolismo , Colorantes Fluorescentes/química , Luz , Microscopía Fluorescente/métodos , Multimerización de Proteína , Receptores Acoplados a Proteínas G/metabolismo , Humanos , Receptores Acoplados a Proteínas G/química
10.
Minerva Ginecol ; 70(5): 539-548, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30226027

RESUMEN

The gonadotropin hormones, follicle stimulating hormone and luteinizing hormone, are essential for reproduction. They work in concert to control multiple aspects of gonadal function to ultimately produce meiotically competent and fertilizable gametes, provide the optimal endometrial environment and support for implantation and maintain pregnancy via progesterone production throughout the first trimester of pregnancy. These complex and multidimensional functions are mediated via the gonadotropin hormone receptors, luteinizing hormone receptor and follicle stimulating hormone receptor, Class A G protein-coupled receptors (GPCR), which couple to multiple G protein-dependent and independent signal pathways to control these physiological processes. Over the last two decades, a plethora of experimental evidence has shown that GPCRs can associate to form dimers and oligomers. This association provides a means of mediating the diverse functional requirements of a single receptor subtype and for the gonadotropin hormone receptors, has been shown to alter the pharmacology and signal activation profile of these receptors. This review will detail the historical and current evidence detailing the formation of gonadotropin hormone receptor homomers and heteromers. We will discuss the functional insights gained from in vitro and in vivo studies, and the potential impact in modulating reproductive health and disease.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Hormona Luteinizante/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Femenino , Humanos , Embarazo , Multimerización de Proteína , Receptores de Gonadotropina/metabolismo , Reproducción/fisiología , Transducción de Señal/fisiología
11.
Endocrinology ; 147(12): 5865-72, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16959838

RESUMEN

11beta-hydroxysteroid dehydrogenase (11betaHSD) enzymes regulate glucocorticoid availability in target tissues. 11betaHSD1 is the predominant isoenzyme expressed and active in human granulosa-lutein (hGL) cells. This study investigated the effects of pharmacological inhibitors of prostaglandin (PG) synthesis on 11betaHSD1 activities and expression in hGL cells. The consequences for 11betaHSD1 of increasing exposure of hGL cells to PGs, either by treatment with exogenous PGs or by challenging cells with IL-1beta, were also assessed. Suppression of basal PG synthesis using four different inhibitors of PG H synthase enzymes [indomethacin, niflumic acid, meclofenamic acid (MA) and N-(2-cyclohexyloxy-4-nitorophenyl) methane sulfonamide (NS-398)] each resulted in significant decreases in both cortisol oxidation and cortisone reduction. Both activities of 11betaHSD1 were suppressed by up to 64+/-6% (P<0.05). Over 4 and 24 h, neither MA nor NS-398 affected the expression of 11betaHSD1 protein, suggesting enzyme regulation by PGs at the posttranslational level. When cells were cotreated for 4 h with PGHS inhibitors plus 30 nm PGD2, PGF2alpha, or PGE2, each PG overcame the suppression of cortisol oxidation by indomethacin or MA. Treatment of hGL cells with IL-1beta increased the concentrations of both PGE2 and PGF2alpha, accompanied by a 70+/-25% increase in net cortisol oxidation. All three responses to IL-1beta were abolished when cells were cotreated with MA. These findings suggest a role for PGs in the posttranslational regulation of 11betaHSD1 activities in hGL cells.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasas/metabolismo , Células Lúteas/enzimología , Prostaglandinas/fisiología , Células Cultivadas , Inhibidores de la Ciclooxigenasa/farmacología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , Ácido Meclofenámico/farmacología , Modelos Biológicos , Nitrobencenos/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Prostaglandinas/biosíntesis , Prostaglandinas/farmacología , Sulfonamidas/farmacología
12.
Methods Cell Biol ; 132: 55-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26928539

RESUMEN

The organization of G protein-coupled receptors (GPCRs) into dimers and higher-order oligomers has provided a potential mechanistic system in defining complex GPCR responses. Despite being studied for nearly 20 years it has, and still is, been an area of controversy. Although technology has developed to quantitatively measure these associations in real time, identify the structural interfaces and even systems to understand the physiological significance of di/oligomerization, key questions remain outstanding including the role of each individual complex from the monomer to the higher-order oligomer, in their native system. Recently, single-molecule microscopy approaches have provided the tools to directly visualize individual GPCRs in dimers and oligomers, though as with any technological development each have their advantages and limitations. This chapter will describe these recent developments in single-molecule fluorescent microscopy, focusing on our recent application of super-resolution imaging of the GPCR for the luteinizing hormone/chorionic gonadotropin to quantify GPCR monomers and formation of protomers in to dimers and distinct oligomeric forms. We present our approach, considerations, strategy, and challenges to visualize this receptor beyond the light diffraction limit via photoactivated localization microscopy with photoactivatable dyes. The addition of super-resolution approaches to the GPCR "nano-tool kit" will pave the way for novel avenues to answer outstanding questions regarding the existence and significance of these complexes to GPCR signaling.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Humanos , Microscopía Fluorescente , Unión Proteica , Multimerización de Proteína , Receptores Acoplados a Proteínas G/ultraestructura , Transducción de Señal , Análisis de la Célula Individual
13.
Methods Cell Biol ; 117: 433-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24143991

RESUMEN

G protein-coupled receptors (GPCRs) mediate a diverse range of physiological functions via activation of complex signaling systems. Organization of GPCRs in to dimers and oligomers provides a mechanism for both signal diversity and specificity in cellular responses, yet our understanding of the physiological significance of dimerization, particularly homodimerization, has not been forthcoming. This chapter will describe how we have investigated the physiological importance of GPCR homodimerization, using the luteinizing hormone/chorionic gonadotropin receptor as a model GPCR. Using transactivation as a mode of assessing receptor dimerization, we describe our cellular system and functional assays for assessment of transactivation in vitro and detail our strategy for generating a mouse model to assess GPCR transactivation in vivo.


Asunto(s)
Fertilidad/genética , Receptores de HL/metabolismo , Testículo/metabolismo , Activación Transcripcional , Animales , Cromosomas Artificiales Bacterianos , Embrión de Mamíferos , Femenino , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Células HEK293 , Humanos , Luciferasas/química , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerización de Proteína , Receptores de HL/química , Receptores de HL/genética , Transducción de Señal , Testículo/citología , Testículo/embriología , Transfección
14.
J Endocrinol ; 203(2): 215-29, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19666697

RESUMEN

In the pituitary, C-type natriuretic peptide (CNP) has been implicated as a gonadotroph-specific factor, yet expression of the CNP gene (Nppc) and CNP activity in gonadotrophs is poorly defined. Here, we examine the molecular expression and putative function of a local gonadotroph natriuretic peptide system. Nppc, along with all three natriuretic peptide receptors (Npr1, Npr2 and Npr3), was expressed in both alphaT3-1 and LbetaT2 cells and primary mouse pituitary tissue, yet the genes for atrial-(ANP) and B-type natriuretic peptides (Nppa and Nppb) were much less abundant. Putative processing enzymes of CNP were also expressed in alphaT3-1 cells and primary mouse pituitaries. Transcriptional analyses revealed that the proximal 50 bp of the murine Nppc promoter were sufficient for GNRH responsiveness, in an apparent protein kinase C and calcium-dependent manner. Electrophoretic mobility shift assays showed Sp1/Sp3 proteins form major complexes within this region of the Nppc promoter. CNP protein was detectable in rat anterior pituitaries, and electron microscopy detected CNP immunoreactivity in secretory granules of gonadotroph cells. Pharmacological analyses of natriuretic peptide receptor activity clearly showed ANP and CNP are potent activators of cGMP production. However, functional studies failed to reveal a role for CNP in regulating cell proliferation or LH secretion. Surprisingly, CNP potently stimulated the human glycoprotein hormone alpha-subunit promoter in LbetaT2 cells but not in alphaT3-1 cells. Collectively, these findings support a role for CNP as the major natriuretic peptide of the anterior pituitary, and for gonadotroph cells as the major source of CNP expression and site of action.


Asunto(s)
Gonadotrofos/metabolismo , Péptido Natriurético Tipo-C/fisiología , Hipófisis/metabolismo , Animales , Células Cultivadas , GMP Cíclico/metabolismo , Femenino , Regulación de la Expresión Génica , Hormonas Glicoproteicas de Subunidad alfa/genética , Hormona Luteinizante/metabolismo , Masculino , Ratones , Péptido Natriurético Tipo-C/análisis , Péptido Natriurético Tipo-C/genética , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores del Factor Natriurético Atrial/análisis , Receptores del Factor Natriurético Atrial/genética
15.
Biol Reprod ; 68(6): 2157-63, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12606327

RESUMEN

In the ovary, cortisol is oxidized to cortisone by 11beta-hydroxysteroid dehydrogenase (11betaHSD). The present study investigated whether follicular fluid (FF) from large antral follicles and spontaneous ovarian cysts, isolated from bovine and porcine ovaries, contained modulators of 11betaHSD activity. Whereas FF from antral follicles had no significant effect over 1 h on NADP+-dependent 11betaHSD activity in rat kidney homogenates, enzyme activity was inhibited by FF from bovine and porcine ovarian cysts (80.5% +/- 2.3% and 72.8% +/- 3.4% of control, respectively). Following C18 reverse-phase chromatography, the hydrophilic fractions of FF from bovine and porcine antral follicles stimulated NADP+-dependent 11betaHSD activities (111.5% +/- 21.6% and 55.2% +/- 5.7% respectively). Hydrophobic compounds inhibited NADP+-dependent cortisol oxidation by 58.2% +/- 5.1% (bovine) and 45.7% +/- 2.0% (porcine). In both species, FF from ovarian cysts appeared to contain less of the hydrophilic stimuli to 11betaHSD activity and more of the hydrophobic inhibitors. The FF from antral follicles and ovarian cysts, and the C18 fractions thereof, had no significant effect on NAD+-dependent cortisol oxidation. The ovarian modulators of NADP+-dependent 11betaHSD activities did not coelute with cortisol, cortisone, estradiol, testosterone, progesterone, pregnenolone, and cholesterol. However, the 11betaHSD stimuli in porcine FF from both antral follicles and cysts coeluted with prostaglandin (PG) E2 and PGF2alpha. We conclude that large antral follicles and spontaneous ovarian cysts, in both the cow and the pig, contain ovarian modulators of the NADP+-dependent 11betaHSD activity. Moreover, FF from spontaneous ovarian cysts, because of decreased content of the 11betaHSD stimulus accompanied by increased content of the 11betaHSD inhibitors, exerts a net inhibitory effect on 11betaHSD activity.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasas/metabolismo , Líquido Folicular/enzimología , Quistes Ováricos/enzimología , Folículo Ovárico/enzimología , Ovario/fisiología , Animales , Bovinos , Femenino , Hormonas Esteroides Gonadales/fisiología , Hidrocortisona/metabolismo , Riñón/enzimología , NAD/fisiología , NADP/fisiología , Oxidación-Reducción , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA