Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(16)2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37629102

RESUMEN

Human patients with mutations within NPPC or NPR2 genes (encoding C-type natriuretic peptide (CNP) and guanylyl cyclase-B (GC-B), respectively) display clinical signs associated with skeletal abnormalities, such as overgrowth or short stature. Mice with induced models of Nppc or Npr2 deletion display profound achondroplasia, dwarfism and early death. Recent pharmacological therapies to treat short stature are utilizing long-acting CNP analogues, but the effects of manipulating CNP expression during development remain unknown. Here, we use Danio rerio (zebrafish) as a model for vertebrate development, employing both pharmacological and reverse genetics approaches to alter expression of genes encoding CNP in zebrafish. Four orthologues of CNP were identified in zebrafish, and spatiotemporal expression profiling confirmed their presence during development. Bioinformatic analyses suggested that nppcl is the most likely the orthologue of mammalian CNP. Exogenous CNP treatment of developing zebrafish embryos resulted in impaired growth characteristics, such as body length, head width and eye diameter. This reduced growth was potentially caused by increased apoptosis following CNP treatment. Expression of endogenous nppcl was downregulated in these CNP-treated embryos, suggesting that negative feedback of the CNP system might influence growth during development. CRISPR knock-down of endogenous nppcl in developing zebrafish embryos also resulted in impaired growth characteristics. Collectively, these data suggest that CNP in zebrafish is crucial for normal embryonic development, specifically with regard to growth.


Asunto(s)
Acondroplasia , Péptido Natriurético Tipo-C , Femenino , Embarazo , Humanos , Animales , Ratones , Péptido Natriurético Tipo-C/genética , Pez Cebra/genética , Trastornos del Crecimiento , Mamíferos
2.
Int J Mol Sci ; 22(3)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499110

RESUMEN

Patients harbouring mutations in genes encoding C-type natriuretic peptide (CNP; NPPC) or its receptor guanylyl cyclase B (GC-B, NPR2) suffer from severe growth phenotypes; loss-of-function mutations cause achondroplasia, whereas gain-of-function mutations cause skeletal overgrowth. Although most of the effects of CNP/GC-B on growth are mediated directly on bone, evidence suggests the natriuretic peptides may also affect anterior pituitary control of growth. Our previous studies described the expression of NPPC and NPR2 in a range of human pituitary tumours, normal human pituitary, and normal fetal human pituitary. However, the natriuretic peptide system in somatotropes has not been extensively explored. Here, we examine the expression and function of the CNP/GC-B system in rat GH3 somatolactotrope cell line and pituitary tumours from a cohort of feline hypersomatotropism (HST; acromegaly) patients. Using multiplex RT-qPCR, all three natriuretic peptides and their receptors were detected in GH3 cells. The expression of Nppc was significantly enhanced following treatment with either 100 nM TRH or 10 µM forskolin, yet only Npr1 expression was sensitive to forskolin stimulation; the effects of forskolin and TRH on Nppc expression were PKA- and MAPK-dependent, respectively. CNP stimulation of GH3 somatolactotropes significantly inhibited Esr1, Insr and Lepr expression, but dramatically enhanced cFos expression at the same time point. Oestrogen treatment significantly enhanced expression of Nppa, Nppc, Npr1, and Npr2 in GH3 somatolactotropes, but inhibited CNP-stimulated cGMP accumulation. Finally, transcripts for all three natriuretic peptides and receptors were expressed in feline pituitary tumours from patients with HST. NPPC expression was negatively correlated with pituitary tumour volume and SSTR5 expression, but positively correlated with D2R and GHR expression. Collectively, these data provide mechanisms that control expression and function of CNP in somatolactotrope cells, and identify putative transcriptional targets for CNP action in somatotropes.


Asunto(s)
Mutación , Péptido Natriurético Tipo-C/metabolismo , Neoplasias Hipofisarias/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Acromegalia/metabolismo , Animales , Gatos , Línea Celular , Colforsina/farmacología , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Estrógenos/metabolismo , Femenino , Masculino , Fenotipo , Hipófisis/metabolismo , Ratas , Ratas Wistar , Hormona Liberadora de Tirotropina/farmacología
3.
Vet Surg ; 50(2): 303-311, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32691934

RESUMEN

OBJECTIVE: To determine the incidence, outcome, and risk factors for postattenuation neurological signs (PANS) in cats treated for single congenital portosystemic shunts (CPSS). STUDY DESIGN: Retrospective cohort study. ANIMALS: Cats (n = 50) with a single CPSS. METHODS: Medical records of cats treated by surgical attenuation of a single CPSS between 2003 and 2017 were reviewed for signalment, surgical technique, preoperative management and postoperative clinical outcomes. Binary logistic regression was performed to investigate risk factors for occurrence of PANS and seizures. RESULTS: Congenital portosystemic shunts in 50 cats included 40 extrahepatic and 10 intrahepatic shunts. Postattenuation neurological signs were recorded in 31 (62%) cats and graded as 1 in 10 cats, 2 in nine cats, and 3 in 12 cats. Postattenuation neurological signs included seizures in 11 cats. Five of 31 cats with PANS did not survive to discharge. No association was detected between PANS or seizures and the type of CPSS (intrahepatic or extrahepatic), degree of attenuation, age, or the use of perioperative levetiracetam or hepatic encephalopathy immediately preoperatively. Osmolality at a median 24 hours postoperatively was lower in cats with PANS (P < .049, Wald 3.867, odds ratio [Exp(B)] 0.855, CI 0.732-0.999). CONCLUSION: Postattenuation neurological signs are common complications in cats treated for CPSS. Preoperative levetiracetam did not prevent the occurrence of PANS or seizures. The only risk factor for PANS detected was lower postoperative Osmolality in cats with PANS at 24 hours. CLINICAL SIGNIFICANCE: Postattenuation neurological signs including seizures occur frequently in cats undergoing surgical attenuation of a CPSS. Preoperative levetiracetam did not protect against the development of PANS.


Asunto(s)
Gatos/cirugía , Sistema Porta/anomalías , Complicaciones Posoperatorias/veterinaria , Convulsiones/veterinaria , Malformaciones Vasculares/veterinaria , Animales , Gatos/anomalías , Estudios de Cohortes , Femenino , Incidencia , Masculino , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Factores de Riesgo , Malformaciones Vasculares/cirugía
4.
Reproduction ; 156(4): 313-330, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30306765

RESUMEN

Equine chorionic girdle trophoblast cells play important endocrine and immune functions critical in supporting pregnancy. Very little is known about the genes and pathways that regulate chorionic girdle trophoblast development. Our aim was to identify genes and signalling pathways active in vivo in equine chorionic girdle trophoblast within a critical 7-days window. We exploited the late implantation of the equine conceptus to obtain trophoblast tissue. An Agilent equine 44K microarray was performed using RNA extracted from chorionic girdle and chorion (control) from equine pregnancy days 27, 30, 31 and 34 (n = 5), corresponding to the initiation of chorionic girdle trophoblast proliferation, differentiation and migration. Data were analysed using R packages limma and maSigPro, Ingenuity Pathway Analysis and DAVID and verified using qRT-PCR, promoter analysis, western blotting and migration assays. Microarray analysis showed gene expression (absolute log FC >2, FDR-adjusted P < 0.05) was rapidly and specifically induced in the chorionic girdle between days 27 and 34 (compared to day 27, day 30 = 116, day 31 = 317, day 34 = 781 genes). Pathway analysis identified 35 pathways modulated during chorionic girdle development (e.g. FGF, integrin, Rho GTPases, MAPK) including pathways that have limited description in mammalian trophoblast (e.g. IL-9, CD40 and CD28 signalling). Rho A and ERK/MAPK activity was confirmed as was a role for transcription factor ELF5 in regulation of the CGB promoter. The purity and accessibility of chorionic girdle trophoblast proved to be a powerful resource to identify candidate genes and pathways involved in early equine placental development.


Asunto(s)
Caballos/embriología , Trofoblastos/metabolismo , Animales , Femenino , Expresión Génica , Caballos/metabolismo , Masculino , Placentación , Embarazo , Transducción de Señal , Transcriptoma
5.
Vet Surg ; 47(6): 745-755, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30084495

RESUMEN

OBJECTIVE: To determine the incidence, outcome, and risk factors for postattenuation neurological signs (PANS) and seizures after attenuation of single congenital portosystemic shunts (CPSS) in dogs. STUDY DESIGN: Retrospective cohort study. SAMPLE POPULATION: Dogs (N = 253) with single CPSS. METHODS: Medical records of dogs treated by surgical attenuation of a single CPSS between February 2000 and July 2015 were reviewed for signalment and preoperative and postoperative clinical outcomes, including the occurrence of PANS. Univariable and multivariable binary logistic regression was used to assess risk factors for PANS and for seizures. RESULTS: Twenty-eight (11.1%) dogs developed PANS, including 12 (4.7%) dogs with seizures. Five (17.9%) dogs with PANS did not survive to discharge. Risk factors for PANS included the presence of hepatic encephalopathy (HE) immediately preoperatively (P = .038, odds ratio [OR] 2.704, CI 1.057-6.922) and increasing age (P < .001, OR 1.476, CI 1.223-1.780). Risk factors for seizures included the presence of HE immediately preoperatively (P = .048, OR 3.538, CI 1.013-12.363) and increasing age (P = .009, OR 1.364, CI 1.082-1.720). No association was found between the location of portosystemic shunts (extrahepatic and intrahepatic) and post-operative PANS (P = .532) or seizures (P = .620). Similarly, preemptive administration of levetiracetam did not influence the risk of PANS (P = .991) or seizures (P = .752). CONCLUSION: Preoperative HE and older age in dogs with a CPSS increased the odds of developing PANS and seizures in our population. Preemptive administration of levetiracetam did not protect dogs against the development of PANS or seizures. CLINICAL SIGNIFICANCE: Surgical attenuation of a single CPSS should not be excessively delayed, and surgeons should stabilize the clinical signs of HE before surgery to prevent postoperative PANS and seizures.


Asunto(s)
Enfermedades de los Perros/cirugía , Sistema Porta/cirugía , Complicaciones Posoperatorias/veterinaria , Malformaciones Vasculares/veterinaria , Animales , Estudios de Cohortes , Enfermedades de los Perros/congénito , Perros , Femenino , Incidencia , Masculino , Sistema Porta/anomalías , Derivación Portosistémica Quirúrgica , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Factores de Riesgo , Malformaciones Vasculares/cirugía
6.
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
7.
J Biol Chem ; 289(11): 7873-83, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24482225

RESUMEN

Gonadotropin-releasing hormone (GnRH) is secreted in brief pulses that stimulate synthesis and secretion of pituitary gonadotropin hormones and thereby mediate control of reproduction. It acts via G-protein-coupled receptors to stimulate effectors, including ERK. Information could be encoded in GnRH pulse frequency, width, amplitude, or other features of pulse shape, but the relative importance of these features is unknown. Here we examine this using automated fluorescence microscopy and mathematical modeling, focusing on ERK signaling. The simplest scenario is one in which the system is linear, and response dynamics are relatively fast (compared with the signal dynamics). In this case integrated system output (ERK activation or ERK-driven transcription) will be roughly proportional to integrated input, but we find that this is not the case. Notably, we find that relatively slow response kinetics lead to ERK activity beyond the GnRH pulse, and this reduces sensitivity to pulse width. More generally, we show that the slowing of response kinetics through the signaling cascade creates a system that is robust to pulse width. We, therefore, show how various levels of response kinetics synergize to dictate system sensitivity to different features of pulsatile hormone input. We reveal the mathematical and biochemical basis of a dynamic GnRH signaling system that is robust to changes in pulse amplitude and width but is sensitive to changes in receptor occupancy and frequency, precisely the features that are tightly regulated and exploited to exert physiological control in vivo.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Transporte Activo de Núcleo Celular , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Hormonas/metabolismo , Humanos , Cinética , Luciferasas/metabolismo , Modelos Teóricos , Regiones Promotoras Genéticas , Transducción de Señal , Transcriptoma
8.
J Biol Chem ; 288(29): 21001-21014, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23754287

RESUMEN

Many extracellular signals act via the Raf/MEK/ERK cascade in which kinetics, cell-cell variability, and sensitivity of the ERK response can all influence cell fate. Here we used automated microscopy to explore the effects of ERK-mediated negative feedback on these attributes in cells expressing endogenous ERK or ERK2-GFP reporters. We studied acute rather than chronic stimulation with either epidermal growth factor (ErbB1 activation) or phorbol 12,13-dibutyrate (PKC activation). In unstimulated cells, ERK-mediated negative feedback reduced the population-average and cell-cell variability of the level of activated ppERK and increased its robustness to changes in ERK expression. In stimulated cells, negative feedback (evident between 5 min and 4 h) also reduced average levels and variability of phosphorylated ERK (ppERK) without altering the "gradedness" or sensitivity of the response. Binning cells according to total ERK expression revealed, strikingly, that maximal ppERK responses initially occur at submaximal ERK levels and that this non-monotonic relationship changes to an increasing, monotonic one within 15 min. These phenomena occur in HeLa cells and MCF7 breast cancer cells and in the presence and absence of ERK-mediated negative feedback. They were best modeled assuming distributive (rather than processive) activation. Thus, we have uncovered a novel, time-dependent change in the relationship between total ERK and ppERK levels that persists without negative feedback. This change makes acute response kinetics dependent on ERK level and provides a "gating" or control mechanism in which the interplay between stimulus duration and the distribution of ERK expression across cells could modulate the proportion of cells that respond to stimulation.


Asunto(s)
Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Retroalimentación Fisiológica , Sistema de Señalización de MAP Quinasas , Proteína Quinasa C/metabolismo , Activación Enzimática/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Células HeLa , Humanos , Cinética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células MCF-7 , Microscopía Fluorescente , Modelos Biológicos , Forbol 12,13-Dibutirato/farmacología , Fosforilación/efectos de los fármacos , Factores de Tiempo
9.
Cell Tissue Res ; 355(2): 425-36, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24352806

RESUMEN

The guanylyl cyclases, GC-A and GC-B, are selective receptors for atrial and C-type natriuretic peptides (ANP and CNP, respectively). In the anterior pituitary, CNP and GC-B are major regulators of cGMP production in gonadotropes and yet mouse models of disrupted CNP and GC-B indicate a potential role in growth hormone secretion. In the current study, we investigate the molecular and pharmacological properties of the CNP/GC-B system in somatotrope lineage cells. Primary rat pituitary and GH3 somatolactotropes expressed functional GC-A and GC-B receptors that had similar EC50 properties in terms of cGMP production. Interestingly, GC-B signaling underwent rapid homologous desensitization in a protein phosphatase 2A (PP2A)-dependent manner. Chronic exposure to either CNP or ANP caused a significant down-regulation of both GC-A- and GC-B-dependent cGMP accumulation in a ligand-specific manner. However, this down-regulation was not accompanied by alterations in the sub-cellular localization of these receptors. Heterologous desensitization of GC-B signaling occurred in GH3 cells following exposure to either sphingosine-1-phosphate or thyrotrophin-releasing hormone (TRH). This heterologous desensitization was protein kinase C (PKC)-dependent, as pre-treatment with GF109203X prevented the effect of TRH on CNP/GC-B signaling. Collectively, these data indicate common and distinct properties of particulate guanylyl cyclase receptors in somatotropes and reveal that independent mechanisms of homologous and heterologous desensitization occur involving either PP2A or PKC. Guanylyl cyclase receptors thus represent potential novel therapeutic targets for treating growth-hormone-associated disorders.


Asunto(s)
Lactotrofos/enzimología , Receptores del Factor Natriurético Atrial/metabolismo , Transducción de Señal , Animales , Factor Natriurético Atrial/farmacología , Señalización del Calcio/efectos de los fármacos , Línea Celular , AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Endocitosis/efectos de los fármacos , Lactotrofos/efectos de los fármacos , Ligandos , Ratones , Péptido Natriurético Tipo-C/farmacología , Proteína Quinasa C/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Esfingolípidos/metabolismo , Hormona Liberadora de Tirotropina/metabolismo
10.
Vet Sci ; 11(3)2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38535834

RESUMEN

Congenital portosystemic shunts (CPSS) are vascular anomalies resulting in liver hypoplasia and hepatic insufficiency. Cats with CPSS typically show signs of hepatic encephalopathy associated with increased ammonia, inflammatory cytokines, and oxidative stress. Surgical attenuation of the CPSS results in improved liver function, resolution of clinical signs, and increased portal blood flow. Hepatic gene expression has not previously been investigated in cats with CPSS. Here, we compared the hepatic expression of genes involved in the urea cycle (CPS1, NAGS), angiogenesis (VEGFR2, NPPA, NPR1, NPPC, NPR2, HIF1a), liver regeneration (SERPINB1, HGF, TGFß), and metabolism (FGF21) from a small series of cats (n = 18) with CPSS to that of control cats (n = 10). The expression of TGFß, VEGFR2, HGF, FGF21, and CPS1 was significantly elevated in liver biopsies from cats with CPSS. Cats that could only tolerate partial closure of their CPSS had increased hepatic expression of SERPINB1, HIF1a, and NPR2 compared with those that could tolerate complete ligation. Furthermore, there were no significant correlations between gene expression and pre-operative plasma ammonia concentrations in cats with CPSS. The changes in hepatic gene expression in cats with CPSS are in direct contrast to those seen in dogs with CPSS, suggesting alternative mechanisms may be involved in mediating hepatic changes in cats with CPSS.

11.
Cell Tissue Res ; 352(3): 751-60, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23568656

RESUMEN

In target tissues, cortisol is metabolised by two 11ß-hydroxysteroid dehydrogenase (11ßHSD) isoenzymes, namely 11ßHSD1 and 11ßHSD2, both of which are co-expressed in the boar testis and reproductive tract. The present study has assessed whether cortisol-cortisone metabolism in boar testis and caput epididymidis can be regulated via the gonadotrophin-cAMP signalling pathway. 11ßHSD activities were measured by using a radiometric conversion assay in static tissue culture. In both testis and caput epididymidis, the net reduction of cortisone but not the net oxidation of cortisol, was significantly decreased by luteinising hormone (by 53 ± 20% and 45 ± 9%, respectively, P < 0.05), forskolin (by 60 ± 7% and 57 ± 9%, respectively, P < 0.01) and 8-bromo-cAMP (by 54 ± 4% and 64 ± 1%, respectively, P < 0.01). This suppression of 11-ketosteroid reductase activity in the boar testis by forskolin could be attenuated by the protein kinase A (PKA) inhibitor, H89. Hence, within the boar testis and the caput epididymidis, the local actions of glucocorticoids are modulated by gonadotrophin-cAMP-PKA signalling via their selective effects on the reductase activity of 11ßHSD.


Asunto(s)
AMP Cíclico/metabolismo , Epidídimo/metabolismo , Glucocorticoides/metabolismo , Gonadotropinas/metabolismo , Transducción de Señal , Porcinos/metabolismo , Testículo/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasas/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Células Cultivadas , Colforsina/farmacología , Cortisona/metabolismo , Disección , Epidídimo/citología , Epidídimo/efectos de los fármacos , Hormona Folículo Estimulante/farmacología , Humanos , Hidrocortisona/metabolismo , Hormona Luteinizante/farmacología , Masculino , Transporte de Proteínas/efectos de los fármacos , Radiometría , Receptores de Glucocorticoides/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos
12.
J Biol Chem ; 285(32): 24360-71, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20507982

RESUMEN

Gonadotropin-releasing hormone (GnRH) acts via G-protein-coupled receptors on gonadotrophs to stimulate synthesis and secretion of luteinizing hormone and follicle-stimulating hormone. It is secreted in pulses, and its effects depend on pulse frequency, but decoding mechanisms are unknown. Here we have used an extracellular signal regulated kinase-green fluorescent protein (ERK2-GFP) reporter to monitor GnRH signaling. GnRH caused dose-dependent ERK2-GFP translocation to the nucleus, providing a live-cell readout for activation. Pulsatile GnRH caused dose- and frequency-dependent ERK2-GFP translocation. These responses were rapid and transient, showed only digital tracking, and did not desensitize under any condition tested (dose, frequency, and receptor number varied). We also tested for the effects of cycloheximide (to prevent induction of nuclear-inducible MAPK phosphatases) and used GFP fusions containing ERK mutations (D319N, which prevents docking domain-dependent binding to MAPK phosphatases, and K52R, which prevents catalytic activity). These manipulations had little or no effect on the translocation responses, arguing against a role for MAPK phosphatases or ERK-mediated feedback in shaping ERK activation during pulsatile stimulation. GnRH also caused dose- and frequency-dependent activation of the alpha-gonadotropin subunit-, luteinizing hormone beta-, and follicle-stimulating hormone beta- luciferase reporters, and the latter response was inhibited by ERK1/2 knockdown. Moreover, GnRH caused frequency-dependent activation of an Egr1-luciferase reporter, but the response was proportional to cumulative pulse duration. Our data suggest that frequency decoding is not due to negative feedback shaping ERK signaling in this model.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores LHRH/metabolismo , Catálisis , Cicloheximida/farmacología , Relación Dosis-Respuesta a Droga , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Procesamiento de Imagen Asistido por Computador , Hormona Luteinizante/metabolismo , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Mutación , Transducción de Señal
13.
J Feline Med Surg ; 23(10): 867-874, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33269622

RESUMEN

OBJECTIVES: The objectives of this study were to validate a commercially available luteinising hormone (LH) cat ELISA, to determine whether the increases in plasma LH concentration that occur after neutering are maintained throughout cats' lives and if other factors such as calendar seasons in both intact and neutered cats, and neutering age in neutered cats, influence plasma LH concentrations. METHODS: Stored plasma samples from client-owned cats were used for the measurement of LH concentrations. Clinical data, including age, sex, age at neutering and medical history, were reviewed. Two populations were included in this study: (1) a senior and geriatric cat population (⩾9 years old), including 18 intact and 18 neutered cats matched for age, sex and month of sample collection; and (2) an adult cat population (2-8 years old), including 45 neutered cats. LH concentrations were measured using a commercially available feline ELISA. RESULTS: Senior and geriatric neutered cats had higher plasma LH concentrations than age-matched intact cats (P <0.001). Calendar season did not influence plasma LH concentrations in the adult (P = 0.727) or senior/geriatric (P = 0.745) cats included in this study. No influence of age at neutering was observed on plasma LH concentrations (P = 0.296). CONCLUSIONS AND RELEVANCE: Neutering causes a significant long-term increase in LH concentrations in cats and further studies are required to determine the consequences on feline health.


Asunto(s)
Hormona Luteinizante , Animales , Gatos
14.
J Feline Med Surg ; 23(2): 131-137, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32684121

RESUMEN

OBJECTIVES: An affordable and effective treatment is needed to manage feline hypersomatotropism. The aim of this study was to assess whether treatment with oral cabergoline for 90 days in cats with hypersomatotropism and diabetes mellitus improved diabetic and insulin-like growth factor 1 (IGF-1) control. METHODS: This was a prospective cohort non-blinded pilot study enrolling client-owned cats with spontaneously occurring diabetes mellitus and hypersomatotropism. Cats received oral cabergoline (5-10 µg/kg q24h) for 90 consecutive days. Serum IGF-1 and fructosamine concentrations were measured on days 1, 30 and 90. Quality of life was determined using the DIAQoL-pet questionnaire on days 1 and 90. RESULTS: Nine cats were enrolled and eight completed the study. There was no significant change in the following: IGF-1 (day 1 median 2001 ng/ml [range 890-2001 ng/ml]; day 30 median 2001 ng/ml [range 929-2001 ng/ml]; day 90 median 1828 ng/ml [range 1035-2001 ng/ml]; χ2(2) = 0.667, P = 0.805); fructosamine (day 1 median 499 µmol/l [range 330-887 µmol/l], day 30 median 551 µmol/l [range 288-722 µmol/l], day 90 median 503 [range 315-851 µmol/l]; χ2(2) = 0.581, P = 0.764); or DIAQoL-pet score (median on day 1 -2.79 [range -4.62 to -0.28], median on day 90 -3.24 [range -4.41 to -0.28]; P = 0.715). There was a significant change of insulin dose (χ2(2) = 8.667, P = 0.008) with cats receiving higher insulin doses at day 90 compared with day 1 (median on day 1 was 0.98 [range 0.63-1.49] and median on day 90 was 1.56 [range 0.49-2.55] units/kg q12h; P = 0.026). CONCLUSIONS AND RELEVANCE: Cabergoline did not improve diabetic control or normalise insulin-like growth factor concentration, or improve patient quality of life.


Asunto(s)
Acromegalia , Enfermedades de los Gatos , Diabetes Mellitus , Acromegalia/veterinaria , Animales , Cabergolina , Enfermedades de los Gatos/tratamiento farmacológico , Gatos , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/veterinaria , Factor I del Crecimiento Similar a la Insulina , Proyectos Piloto , Estudios Prospectivos , Calidad de Vida
15.
J Vet Intern Med ; 35(2): 823-833, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33624865

RESUMEN

BACKGROUND: Hypersomatotropism (HST) is an increasingly recognized endocrinopathy in cats and is mostly described associated with diabetes mellitus (DM). OBJECTIVES: To evaluate the efficacy and safety of transsphenoidal hypophysectomy in treating HST and DM in cats. ANIMALS: Sixty-eight client-owned cats with HST and DM treated by transsphenoidal hypophysectomy. METHODS: Retrospective cohort study. Medical records were reviewed for glycemic control and serum insulin-like growth factor-1 (IGF-1) concentrations. Postoperative complications, death within 4 weeks, and proportion achieving diabetic remission were recorded. Survival times and DM-free intervals were calculated. RESULTS: Fifty-eight cats (85.3%) were alive 4 weeks postoperatively with 10 (15%) postoperative deaths. Complications included hypoglycemia (n = 9), electrolyte imbalance (n = 9), and transient congestive heart failure (n = 5). Fifty-five cats (95% of 58 surviving cats [81% of all cats undergoing surgery]) had improved control of diabetes. Diabetic remission occurred in 41 cats (71% of 58 surviving cats [60% of all cats]) with insulin administration discontinued after a median of 9 days (range, 2-120). Postoperative 4-week serum IGF-1 concentration nadir was significantly lower in cats achieving diabetic remission (median 20 ng/mL [15-708] than those that did not (324 ng/mL [15-1955]; P = .03). All cats received long-term levothyroxine and hydrocortisone PO, alongside desmopressin (conjunctival) in 38 of 53 cats (72%). Recurrence of DM occurred in 5 of 41 cats (12%) after a median of 248 days (range, 84-1232). Median survival time of all cats was 853 days (range, 1-1740). CONCLUSIONS AND CLINICAL IMPORTANCE: Transsphenoidal hypophysectomy is an effective treatment for cats with HST and DM, with a long-term outcome that compares favorably to existing options.


Asunto(s)
Acromegalia , Enfermedades de los Gatos , Diabetes Mellitus , Acromegalia/veterinaria , Animales , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/etiología , Enfermedades de los Gatos/cirugía , Gatos , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/veterinaria , Hipofisectomía/veterinaria , Insulina/uso terapéutico , Estudios Retrospectivos
16.
Cells ; 10(2)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33672024

RESUMEN

C-type natriuretic peptide (CNP) is the major natriuretic peptide of the central nervous system and acts via its selective guanylyl cyclase-B (GC-B) receptor to regulate cGMP production in neurons, astrocytes and endothelial cells. CNP is implicated in the regulation of neurogenesis, axonal bifurcation, as well as learning and memory. Several neurological disorders result in toxic concentrations of ammonia (hyperammonaemia), which can adversely affect astrocyte function. However, the relationship between CNP and hyperammonaemia is poorly understood. Here, we examine the molecular and pharmacological control of CNP in rat C6 glioma cells and rat GPNT brain endothelial cells, under conditions of hyperammonaemia. Concentration-dependent inhibition of C6 glioma cell proliferation by hyperammonaemia was unaffected by CNP co-treatment. Furthermore, hyperammonaemia pre-treatment (for 1 h and 24 h) caused a significant inhibition in subsequent CNP-stimulated cGMP accumulation in both C6 and GPNT cells, whereas nitric-oxide-dependent cGMP accumulation was not affected. CNP-stimulated cGMP efflux from C6 glioma cells was significantly reduced under conditions of hyperammonaemia, potentially via a mechanism involving changed in phosphodiesterase expression. Hyperammonaemia-stimulated ROS production was unaffected by CNP but enhanced by a nitric oxide donor in C6 cells. Extracellular vesicle production from C6 cells was enhanced by hyperammonaemia, and these vesicles caused impaired CNP-stimulated cGMP signalling in GPNT cells. Collectively, these data demonstrate functional interaction between CNP signalling and hyperammonaemia in C6 glioma and GPNT cells, but the exact mechanisms remain to be established.


Asunto(s)
GMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Glioma/metabolismo , Hiperamonemia/metabolismo , Animales , Encéfalo/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Péptidos Natriuréticos/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
17.
J Biol Chem ; 284(51): 35746-57, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19858197

RESUMEN

Gonadotropin-releasing hormone (GnRH) acts via 7 transmembrane region receptors on gonadotrophs to stimulate synthesis and secretion of the luteinizing hormone and follicle-stimulating hormone. It is secreted in pulses, and its effects depend on pulse frequency, but decoding mechanisms are unknown. Here we have used (nuclear factor of activated T-cells 2 (NFAT2)-emerald fluorescent protein) to monitor GnRH signaling. Increasing [Ca(2+)](i) causes calmodulin/calcineurin-dependent nuclear NFAT translocation, a response involving proteins (calmodulins and NFATs) that decode frequency in other systems. Using live cell imaging, pulsatile GnRH caused dose- and frequency-dependent increases in nuclear NFAT2-emerald fluorescent protein, and at low frequency, translocation simply tracked GnRH exposure (albeit with slower kinetics). At high frequency (30-min intervals), failure to return to basal conditions before repeat stimulation caused integrative tracking, illustrating how the relative dynamics of up- and downstream signals can increase efficiency of GnRH action. Mathematical modeling predicted desensitization of GnRH effects on [Ca(2+)](i) and that desensitization would increase with dose, frequency, and receptor number, but no such desensitization was seen in HeLa and/or LbetaT2 cells possibly because pulsatile GnRH did not reduce receptor expression (measured by immunofluorescence). GnRH also caused dose- and frequency-dependent activation of alphaGSU, luteinizing hormone beta, and follicle-stimulating hormone beta luciferase reporters, effects that were blocked by calcineurin inhibition. Pulsatile GnRH also activated an NFAT-responsive luciferase reporter, but this response was directly related to cumulative pulse duration. This together with the lack of desensitization of translocation responses suggests that NFAT may mediate GnRH action but is not a genuine decoder of GnRH pulse frequency.


Asunto(s)
Relojes Biológicos/fisiología , Señalización del Calcio/fisiología , Núcleo Celular/metabolismo , Modelos Biológicos , Factores de Transcripción NFATC/metabolismo , Receptores LHRH/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Animales , Relojes Biológicos/efectos de los fármacos , Calcineurina/genética , Calcineurina/metabolismo , Señalización del Calcio/efectos de los fármacos , Calmodulina/genética , Calmodulina/metabolismo , Núcleo Celular/genética , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Células HeLa , Humanos , Ratones , Factores de Transcripción NFATC/genética , Receptores LHRH/genética
18.
Basic Res Cardiol ; 105(2): 257-66, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19714395

RESUMEN

We demonstrated previously that adrenomedullin (AM), when given during early reperfusion, limited infarct size in rat heart. The present study was undertaken to provide direct evidence of the NO-dependency of AM's cardioprotective action by assessing NO biosynthesis and involvement of the soluble guanylyl cyclase (sGC) pathway. Perfused hearts from male CD-1 mice were subjected to 30-min left coronary occlusion and 60-min reperfusion. Infarct size was determined by tetrazolium staining. AM 10 nM was administered from 20 min after coronary occlusion until 10 min after reperfusion. Coronary effluent was analysed for NO2- and NO3-, and myocardial samples were analysed for NO2-, NO3-, nitroso-adducts and cGMP concentration. To examine the role of NO/sGC signalling in the infarct-limiting action of AM, further hearts received the sGC inhibitor ODQ 2 microM. AM treatment stimulated NO synthesis, indicated by increased NO2- efflux in coronary effluent throughout reperfusion (summarised as area under curve, AM 29.2 +/- 3.9 vs. control 14.4 +/- 2.8 micromol min2 mL(-1), P < 0.05). AM limited infarct size (35.4 +/- 2.7 vs. 12.2 +/- 2.3%, P < 0.01), associated with a 2.45-fold increase (P < 0.05) in myocardial cGMP concentration at 10 min after reperfusion. ODQ abolished the infarct size-limiting effect of AM (28.9 +/- 4.3%). These data provide the first evidence that AM increases NO bioavailability in intact murine myocardium and confirm that the NO/sGC/cGMP pathway is central to the cytoprotective action of AM against ischaemia-reperfusion injury.


Asunto(s)
Adrenomedulina/metabolismo , GMP Cíclico/metabolismo , Guanilato Ciclasa/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico/biosíntesis , Adrenomedulina/administración & dosificación , Animales , Técnicas In Vitro , Masculino , Ratones , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Transducción de Señal
19.
Cell Biochem Funct ; 28(2): 95-106, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20104507

RESUMEN

Since the discovery of endogenously-produced hydrogen sulfide (H(2)S) in various tissues, there has been an explosion of interest in H(2)S as a biological mediator alongside other gaseous mediators, nitric oxide and carbon monoxide. The identification of enzyme-regulated H(2)S synthetic pathways in the cardiovascular system has led to a number of studies examining specific regulatory actions of H(2)S. We review evidence showing that endogenously-generated and exogenously-administered H(2)S exerts a wide range of actions in vascular and myocardial cells including vasodilator/vasoconstrictor effects via modification of the smooth muscle tone, induction of apoptosis and anti-proliferative responses in the smooth muscle cells, angiogenic actions, effects relevant to inflammation and shock, and cytoprotection in models of myocardial ischemia-reperfusion injury. Several molecular mechanisms of action of H(2)S have been described. These include interactions of H(2)S with NO, redox regulation of multiple signaling proteins and regulation of K(ATP) channel opening. The gaps in our current understanding of precise mechanisms, the absence of selective pharmacological tools and the limited availability of H(2)S measurement techniques for living tissues, leave many questions about physiological and pathophysiological roles of H(2)S unanswered at present. Nevertheless, this area of investigation is advancing rapidly. We believe H(2)S holds promise as an endogenous mediator controlling a wide range of cardiovascular cell functions and integrated responses under both physiological and pathological conditions and may be amenable to therapeutic manipulation.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Presión Sanguínea , Bovinos , Humanos , Mediadores de Inflamación/metabolismo , Canales KATP/metabolismo , Músculo Liso Vascular/citología , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico/metabolismo , Ratas
20.
Animals (Basel) ; 10(4)2020 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-32244408

RESUMEN

Automatic milking systems (AMS) are a low-labour alternative to conventional parlours, with previous studies demonstrating that cows vary in their ability to cope with the change to AMS. Cortisol expression can be combined with other measures to assess stress: saliva and hair have the advantage of requiring minimally invasive sampling. No work has investigated the long-term impact of introduction of AMS. The aims of the study were to assess short-term and chronic stress associated with a change in milking system by measuring salivary and hair cortisol levels and to assess the impact on health and production parameters. Cows from one farm changing their milking system were recruited to the study and sampled for saliva (n = 10) and hair (n = 12) before and after installation. Cortisol levels were measured using a salivary cortisol enzyme immunoassay kit. Body condition, lameness and milk parameters of the whole herd were regularly assessed. Salivary cortisol showed no diurnal pattern but was affected by lameness and gestation. Non-lame cows showed a reduction in salivary cortisol after AMS introduction (p < 0.001). Hair cortisol levels increased after AMS, but it was unclear if this change was seasonal. Milk yield increased by 13% and somatic cell count reduced by 28%. Body condition score was consistently good, but lameness remained high throughout the study. Production values alone do not represent high welfare. The high lameness and associated cortisol levels suggest that cow stress requires consideration when changing milking systems.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA