Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 376(1): 51-70, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30467710

RESUMEN

We have previously shown that the chromogranin A (CgA)-derived peptide catestatin (CST: hCgA352-372) inhibits nicotine-induced secretion of catecholamines from the adrenal medulla and chromaffin cells. In the present study, we seek to determine whether CST regulates dense core (DC) vesicle (DCV) quanta (catecholamine and chromogranin/secretogranin proteins) during acute (0.5-h treatment) or chronic (24-h treatment) cholinergic (nicotine) or peptidergic (PACAP, pituitary adenylyl cyclase activating polypeptide) stimulation of PC12 cells. In acute experiments, we found that both nicotine (60 µM) and PACAP (0.1 µM) decreased intracellular norepinephrine (NE) content and increased 3H-NE secretion, with both effects markedly inhibited by co-treatment with CST (2 µM). In chronic experiments, we found that nicotine and PACAP both reduced DCV and DC diameters and that this effect was likewise prevented by CST. Nicotine or CST alone increased expression of CgA protein and together elicited an additional increase in CgA protein, implying that nicotine and CST utilize separate signaling pathways to activate CgA expression. In contrast, PACAP increased expression of CgB and SgII proteins, with a further potentiation by CST. CST augmented the expression of tyrosine hydroxylase (TH) but did not increase intracellular NE levels, presumably due to its inability to cause post-translational activation of TH through serine phosphorylation. Co-treatment of CST with nicotine or PACAP increased quantal size, plausibly due to increased synthesis of CgA, CgB and SgII by CST. We conclude that CST regulates DCV quanta by acutely inhibiting catecholamine secretion and chronically increasing expression of CgA after nicotinic stimulation and CgB and SgII after PACAPergic stimulation.


Asunto(s)
Catecolaminas/metabolismo , Cromogranina A/fisiología , Cromograninas/metabolismo , Nicotina/farmacología , Fragmentos de Péptidos/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Animales , Cromogranina A/farmacología , Hormonas Glicoproteicas de Subunidad alfa/metabolismo , Humanos , Norepinefrina/metabolismo , Células PC12 , Fragmentos de Péptidos/farmacología , Ratas , Proteínas de Secreción de la Vesícula Seminal/metabolismo , Transducción de Señal/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
2.
FASEB J ; 32(9): 4815-4823, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29913555

RESUMEN

The pathogenesis of diabetic neuropathy remains enigmatic. Damage to the vasa nervorum may be responsible for this disorder. Recently, we showed that secretoneurin (SN) induces angiogenesis in hindlimb and myocardial ischemia. Moreover, beneficial effects were observed in wound healing. We therefore hypothesized that SN therapy may ameliorate diabetic neuropathy. We used db/db mice as animal model for neuropathy. Gene therapy was accomplished by intramuscular injection of SN plasmid along the sciatic nerve. Sciatic nerve motor and sensory conduction velocities were then measured for 9 wk. Nerve conduction velocities showed normal values in heterozygous mice for the observational period, but were severely reduced in homozygous mice in which velocities were significantly improved by SN, but not by control plasmid gene therapy. The reaction time in the tail-flick test improved significantly in SN-treated animals. The induction of growth of vasa nervorum seems to be part of the underlying mechanism. In addition, SN positively affected Schwann cell function in vitro and induced activation of important signaling pathways. Our observations suggest that SN exerts beneficial effects on nerve function in vivo and on Schwann cells in vitro. It therefore may be a promising treatment option for diabetic neuropathy.-Theurl, M., Lener, D., Albrecht-Schgoer, K., Beer, A., Schgoer, W., Liu, Y., Stanzl, U., Fischer-Colbrie, R., Kirchmair, R. Gene therapy with the angiogenic neuropeptide secretoneurin ameliorates experimental diabetic neuropathy.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Neuropatías Diabéticas/terapia , Terapia Genética , Neuropéptidos/uso terapéutico , Secretogranina II/uso terapéutico , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Neuropatías Diabéticas/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Neovascularización Fisiológica/genética , Neuropéptidos/metabolismo , Células de Schwann/metabolismo , Secretogranina II/metabolismo
3.
Circulation ; 126(21): 2491-501, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23081990

RESUMEN

BACKGROUND: Secretoneurin is a neuropeptide located in nerve fibers along blood vessels, is upregulated by hypoxia, and induces angiogenesis. We tested the hypothesis that secretoneurin gene therapy exerts beneficial effects in a rat model of myocardial infarction and evaluated the mechanism of action on coronary endothelial cells. METHODS AND RESULTS: In vivo secretoneurin improved left ventricular function, inhibited remodeling, and reduced scar formation. In the infarct border zone, secretoneurin induced coronary angiogenesis, as shown by increased density of capillaries and arteries. In vitro secretoneurin induced capillary tubes, stimulated proliferation, inhibited apoptosis, and activated Akt and extracellular signal-regulated kinase in coronary endothelial cells. Effects were abrogated by a vascular endothelial growth factor (VEGF) antibody, and secretoneurin stimulated VEGF receptors in these cells. Secretoneurin furthermore increased binding of VEGF to endothelial cells, and binding was blocked by heparinase, indicating that secretoneurin stimulates binding of VEGF to heparan sulfate proteoglycan binding sites. Additionally, secretoneurin increased binding of VEGF to its coreceptor neuropilin-1. In endothelial cells, secretoneurin also stimulated fibroblast growth factor receptor-3 and insulin-like growth factor-1 receptor, and in coronary vascular smooth muscle cells, we observed stimulation of VEGF receptor-1 and fibroblast growth factor receptor-3. Exposure of cardiac myocytes to hypoxia and ischemic heart after myocardial infarction revealed increased secretoneurin messenger RNA and protein. CONCLUSIONS: Our data show that secretoneurin acts as an endogenous stimulator of VEGF signaling in coronary endothelial cells by enhancing binding of VEGF to low-affinity binding sites and neuropilin-1 and stimulates further growth factor receptors like fibroblast growth factor receptor-3. Our in vivo findings indicate that secretoneurin may be a promising therapeutic tool in ischemic heart disease.


Asunto(s)
Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Neuropéptidos/administración & dosificación , Secretogranina II/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Terapia Genética/métodos , Humanos , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica/fisiología , Neuropéptidos/genética , Plásmidos/administración & dosificación , Plásmidos/genética , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , Ratas , Secretogranina II/genética , Transducción de Señal/fisiología
4.
Mamm Genome ; 24(7-8): 276-85, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23839232

RESUMEN

Genes subjected to genomic imprinting are often associated with prenatal and postnatal growth. Furthermore, it has been observed that maternally silenced/paternally expressed genes tend to favour offspring growth, whilst paternally silenced/maternally expressed genes will restrict growth. One imprinted cluster in which this has been shown to hold true is the Gnas cluster; of the three proteins expressed from this cluster, two, Gsα and XLαs, have been found to affect postnatal growth in a number of different mouse models. The remaining protein in this cluster, NESP55, has not yet been shown to be involved in growth. We previously described a new mutation, Ex1A-T, which upon paternal transmission resulted in postnatal growth retardation due to loss of imprinting of Gsα and loss of expression of the paternally expressed XLαs. Here we describe maternal inheritance of Ex1A-T which gives rise to a small but highly significant overgrowth phenotype which we attribute to reduction of maternally expressed NESP55.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Patrón de Herencia/genética , Animales , Tamaño Corporal/genética , Densidad Ósea/genética , Cromograninas , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Impresión Genómica/genética , Masculino , Ratones , Ratones Transgénicos , Fenotipo
5.
Am J Physiol Endocrinol Metab ; 301(2): E288-97, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21521715

RESUMEN

Secretoneurin (SN) is a functional secretogranin II (SgII)-derived peptide that stimulates luteinizing hormone (LH) production and its release in the goldfish. However, the effects of SN on the pituitary of mammalian species and the underlying mechanisms remain poorly understood. To study SN in mammals, we adopted the mouse LßT2 gonadotropin cell line that has characteristics consistent with normal pituitary gonadotrophs. Using radioimmunoassay and real-time RT-PCR, we demonstrated that static treatment with SN induced a significant increment of LH release and production in LßT2 cells in vitro. We found that GnRH increased cellular SgII mRNA level and total SN-immunoreactive protein release into the culture medium. We also report that SN activated the extracellular signal-regulated kinases (ERK) in either 10-min acute stimulation or 3-h chronic treatment. The SN-induced ERK activation was significantly blocked by pharmacological inhibition of MAPK kinase (MEK) with PD-98059 and protein kinase C (PKC) with bisindolylmaleimide. SN also increased the total cyclic adenosine monophosphate (cAMP) levels similarly to GnRH. However, SN did not activate the GnRH receptor. These data indicate that SN activates the protein kinase A (PKA) and cAMP-induced ERK signaling pathways in the LH-secreting mouse LßT2 pituitary cell line.


Asunto(s)
Gonadotrofos/fisiología , Hormona Luteinizante de Subunidad beta/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Neuropéptidos/genética , Secretogranina II/genética , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Cromogranina A/genética , Cromogranina A/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Carpa Dorada , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Células HEK293 , Humanos , Indoles/metabolismo , Hormona Luteinizante de Subunidad beta/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Maleimidas/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Neuropéptidos/inmunología , Neuropéptidos/farmacología , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Secretogranina II/inmunología , Secretogranina II/farmacología
6.
Circ Res ; 105(10): 994-1002, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19797703

RESUMEN

RATIONALE: The neuropeptide secretoneurin induces angiogenesis and postnatal vasculogenesis and is upregulated by hypoxia in skeletal muscle cells. OBJECTIVE: We sought to investigate the effects of secretoneurin on therapeutic angiogenesis. METHODS AND RESULTS: We generated a secretoneurin gene therapy vector. In the mouse hindlimb ischemia model secretoneurin gene therapy by intramuscular plasmid injection significantly increased secretoneurin content of injected muscles, improved functional parameters, reduced tissue necrosis, and restored blood perfusion. Increased muscular density of capillaries and arterioles/arteries demonstrates the capability of secretoneurin gene therapy to induce therapeutic angiogenesis and arteriogenesis. Furthermore, recruitment of endothelial progenitor cells was enhanced by secretoneurin gene therapy consistent with induction of postnatal vasculogenesis. Additionally, secretoneurin was able to activate nitric oxide synthase in endothelial cells and inhibition of nitric oxide inhibited secretoneurin-induced effects on chemotaxis and capillary tube formation in vitro. In vivo, secretoneurin induced nitric oxide production and inhibition of nitric oxide attenuated secretoneurin-induced effects on blood perfusion, angiogenesis, arteriogenesis, and vasculogenesis. Secretoneurin also induced upregulation of basic fibroblast growth factor and platelet-derived growth factor-B in endothelial cells. CONCLUSIONS: In summary, our data indicate that gene therapy with secretoneurin induces therapeutic angiogenesis, arteriogenesis, and vasculogenesis in the hindlimb ischemia model by a nitric oxide-dependent mechanism.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Citocinas/biosíntesis , Terapia Genética , Isquemia/terapia , Neovascularización Fisiológica , Neuropéptidos/biosíntesis , Óxido Nítrico/metabolismo , Secretogranina II/biosíntesis , Animales , Citocinas/genética , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Miembro Posterior/metabolismo , Humanos , Isquemia/genética , Isquemia/metabolismo , Ratones , Neuropéptidos/genética , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Secretogranina II/genética , Células Madre/metabolismo
7.
Transl Neurodegener ; 10(1): 37, 2021 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-34565482

RESUMEN

BACKGROUND: New fluid biomarkers for Alzheimer's disease (AD) that reveal synaptic and neural network dysfunctions are needed for clinical practice and therapeutic trial design. Dense core vesicle (DCV) cargos are promising cerebrospinal fluid (CSF) indicators of synaptic failure in AD patients. However, their value as biomarkers has not yet been determined. METHODS: Immunoassays were performed to analyze the secretory proteins prohormone convertases PC1/3 and PC2, carboxypeptidase E (CPE), secretogranins SgIII and SgII, and Cystatin C in the cerebral cortex (n = 45, provided by Bellvitge University Hospital) and CSF samples (n = 66, provided by The Sant Pau Initiative on Neurodegeneration cohort) from AD patients (n = 56) and age-matched controls (n = 55). RESULTS: In AD tissues, most DCV proteins were aberrantly accumulated in dystrophic neurites and activated astrocytes, whereas PC1/3, PC2 and CPE were also specifically accumulated in hippocampal granulovacuolar degeneration bodies. AD individuals displayed an overall decline of secretory proteins in the CSF. Interestingly, in AD patients, the CSF levels of prohormone convertases strongly correlated inversely with those of neurodegeneration markers and directly with cognitive impairment status. CONCLUSIONS: These results demonstrate marked alterations of neuronal-specific prohormone convertases in CSF and cortical tissues of AD patients. The neuronal DCV cargos are biomarker candidates for synaptic dysfunction and neurodegeneration in AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/metabolismo , Biomarcadores/líquido cefalorraquídeo , Corteza Cerebral/metabolismo , Disfunción Cognitiva/líquido cefalorraquídeo , Vesículas de Núcleo Denso , Humanos
8.
J Anat ; 216(5): 611-24, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20525089

RESUMEN

In long bones of murine species, undisturbed development of the epiphysis depends on the generation of vascularized cartilage canals shortly after birth. Despite its importance, it is still under discussion how this event is exactly regulated. It was suggested previously that, following increased hypoxia in the epiphyseal core, angiogenic factors are expressed and hence stimulate the ingrowth of the vascularized canals. In the present study, we tested this model and examined the spatio-temporal distribution of two angiogenic molecules during early development in mice. In addition, we investigated the onset of cartilage hypertrophy and mineralization. Our results provide evidence that the vascular endothelial growth factor is expressed in the epiphyseal resting cartilage prior to the moment of canal formation and is continuously expressed until the establishment of a large secondary ossification centre. Interestingly, we found no expression of secretoneurin before the establishment of the canals although this factor attracts blood vessels under hypoxic conditions. Epiphyseal development further involves maturation of the resting chondrocytes into hypertrophic ones, associated with the mineralization of the cartilage matrix and eventual death of the latter cells. Our results suggest that vascular endothelial growth factor is the critical molecule for the generation of the epiphyseal vascular network in mice long bones. Secretoneurin, however, does not appear to be a player in this event. Hypertrophic chondrocytes undergo cell death by a mechanism interpreted as chondroptosis.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/metabolismo , Cartílago/metabolismo , Epífisis/crecimiento & desarrollo , Epífisis/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Huesos/citología , Cartílago/citología , Ratones , Factores de Tiempo
9.
J Neurochem ; 110(1): 143-56, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19594665

RESUMEN

Vesicular transmitter release from astrocytes influences neuronal development, function and plasticity. However, secretory pathways and the involved molecular mechanisms in astroglial cells are poorly known. In this study, we show that a variety of SNARE and Munc18 isoforms are expressed by cultured astrocytes, with syntaxin-4, Munc18c, SNAP-23 and VAMP-3 being the most abundant variants. Exocytotic protein expression was differentially regulated by activating and differentiating agents. Specifically, proteins controlling Ca(2+)-dependent secretion in neuroendocrine cells were up-regulated after long-term 8Br-cAMP administration in astrocytes, but not by proinflammatory cytokines. Moreover, 8Br-cAMP treatment greatly increased the cellular content of the peptidic vesicle marker secretogranin-2. Release assays performed on cAMP-treated astrocytes showed that basal and stimulated secretogranin-2 secretion are dependent on [Ca(2+)](i). As shown release of the chimeric hormone ANP.emd from transfected cells, cAMP-induced differentiation in astrocytes enhances Ca(2+)-regulated peptide secretion. We conclude that astroglial cells display distinctive molecular components for exocytosis. Moreover, the regulation of both exocytotic protein expression and Ca(2+)-dependent peptide secretion in astrocytes by differentiating and activating agents suggest that glial secretory pathways are adjusted in different physiological states.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio/fisiología , Exocitosis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neurosecreción/fisiología , Péptidos/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Astrocitos/efectos de los fármacos , Encéfalo/citología , Encéfalo/metabolismo , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Cromograninas/efectos de los fármacos , Cromograninas/metabolismo , Perros , Exocitosis/efectos de los fármacos , Ratones , Proteínas Munc18/química , Proteínas Munc18/metabolismo , Proteínas del Tejido Nervioso/química , Neurosecreción/efectos de los fármacos , Ratas , Proteínas SNARE/química , Proteínas SNARE/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Regulación hacia Arriba/fisiología
10.
J Comp Neurol ; 506(4): 733-44, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18067150

RESUMEN

The immunohistochemical expression of a novel chromogranin-like protein, neuroendocrine secretory protein 55 (NESP55), in the rat spinal cord was investigated. NESP55-immunoreactive cells were detected in the ventral horn, intermediate laminae, and deep dorsal horn, comprising motoneurons, autonomic neurons, and interneurons throughout all spinal segments. Within laminae I-II of the dorsal horn, one or two NESP55-positive cells were often seen. Nerve fibers also contained NESP55 immunoreactivity (IR) and were particularly prominent in the ventral horn. No nerve terminals/varicosities appeared to contain NESP55 in any spinal lamina. Double-staining experiments revealed that a high proportion of the NESP55-positive neurons were cholinergic. Moreover, NESP55-IR in the motoneurons was evenly distributed in the whole cytoplasm with a finely granular appearance. In contrast, the fluorescent material in the preganglionic neurons was concentrated in the perinuclear region and largely overlapped with the trans-Golgi network marker TGN38. Our data provide detailed morphological information on the distribution of NESP55-IR in the rat spinal cord. Also, the differential intracellular expression of NESP55-IR in the spinal motoneurons and autonomic neurons suggests that NESP55 may be processed into different secretory granules and may be involved in both constitutive and regulated pathways in these neurons.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Neuronas/metabolismo , Médula Espinal/metabolismo , Acetilcolina/metabolismo , Animales , Sistema Nervioso Autónomo/citología , Sistema Nervioso Autónomo/metabolismo , Axones/metabolismo , Axones/ultraestructura , Cromograninas , Citoplasma/metabolismo , Citoplasma/ultraestructura , Femenino , Inmunohistoquímica , Interneuronas/citología , Interneuronas/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Neuronas/citología , Neurosecreción/fisiología , Ratas , Ratas Sprague-Dawley , Vesículas Secretoras/metabolismo , Vesículas Secretoras/ultraestructura , Médula Espinal/citología
11.
FASEB J ; 21(11): 2906-17, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17504977

RESUMEN

Expression of angiogenic cytokines like vascular endothelial growth factor is enhanced by hypoxia. We tested the hypothesis that decreased oxygen levels up-regulate the angiogenic factor secretoneurin. In vivo, muscle cells of mouse ischemic hind limbs showed increased secretoneurin expression, and inhibition of secretoneurin by a neutralizing antibody impaired the angiogenic response in this ischemia model. In a mouse soft tissue model of hypoxia, secretoneurin was increased in subcutaneous muscle fibers. In vitro, secretoneurin mRNA and protein were up-regulated in L6 myoblast cells after exposure to low oxygen levels. The hypoxia-dependent regulation of secretoneurin was tissue specific and was not observed in endothelial cells, vascular smooth muscle cells, or AtT20 pituitary tumor cells. The hypoxia-dependent induction of secretoneurin in L6 myoblasts is regulated by hypoxia-inducible factor-1alpha, since inhibition of this factor using si-RNA inhibited up-regulation of secretoneurin. Induction of secretoneurin by hypoxia was dependent on basic fibroblast growth factor in vivo and in vitro, and inhibition of this regulation by heparinase suggests an involvement of low-affinity basic fibroblast growth factor binding sites. In summary, our data show that the angiogenic cytokine secretoneurin is up-regulated by hypoxia in muscle cells by hypoxia-inducible factor-1alpha- and basic fibroblast growth factor-dependent mechanisms.


Asunto(s)
Hipoxia de la Célula , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mioblastos/metabolismo , Neuropéptidos/metabolismo , Secretogranina II/metabolismo , Transducción de Señal , Animales , Western Blotting , Células Cultivadas , Cartilla de ADN/química , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Extremidades/cirugía , Técnica del Anticuerpo Fluorescente , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/irrigación sanguínea , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , NAD/metabolismo , Neovascularización Fisiológica , Neoplasias Hipofisarias/irrigación sanguínea , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Reacción en Cadena de la Polimerasa , Proproteína Convertasas/metabolismo , ARN Interferente Pequeño/farmacología , Radioinmunoensayo , Ratas , Piel/metabolismo , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Peptides ; 29(9): 1561-5, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18550223

RESUMEN

By means of highly sensitive radioimmunoassays, the levels of substance P (SP) and secretoneurin (SN) were detected in vitreous aspirates of patients with macular holes which served as controls, in patients with nonproliferative diabetic retinopathy (DR), active proliferative diabetic retinopathy (active PDR), inactive PDR, rhegmatogenous retinal detachment and proliferative vitreoretinopathy (PVR). Furthermore, SN-like immunoreactivities were characterized by reversed phase-HPLC. The concentration of SN was more than 20-fold higher in macular holes when compared with SP and reversed phase HPLC revealed evidence that the vitreous levels of SN represent authentic SN. SN was significantly decreased in patients with nonproliferative DR, active PDR and inactive PDR by more than 70% which seems to result from a reduced expression and/or secretion from the cilary epithelium and a reduced release from the retina both due to diabetes mellitus. By contrast SP was increased in rhegmatogenous retinal detachment most obviously due to an enhanced outflow of the peptide through retinal breaks. Despite their proangiogenic activities, SP and SN are unlikely to be involved in the pathogenesis of neovascularizations in DR because of their unchanged and reduced levels, respectively, but the low levels of both peptides may facilitate the regression of vasoproliferations following laser photocoagulation.


Asunto(s)
Retinopatía Diabética/metabolismo , Neuropéptidos/análisis , Desprendimiento de Retina/metabolismo , Perforaciones de la Retina/metabolismo , Secretogranina II/análisis , Sustancia P/análisis , Vitreorretinopatía Proliferativa/metabolismo , Cuerpo Vítreo/metabolismo , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad
13.
Mol Cell Biol ; 25(8): 3019-26, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15798190

RESUMEN

Genomic imprinting results in parent-of-origin-dependent monoallelic expression of selected genes. Although their importance in development and physiology is recognized, few imprinted genes have been investigated for their effects on brain function. Gnas is a complex imprinted locus whose gene products are involved in early postnatal adaptations and neuroendocrine functions. Gnas encodes the stimulatory G-protein subunit Gsalpha and two other imprinted protein-coding transcripts. Of these, the Nesp transcript, expressed exclusively from the maternal allele, codes for neuroendocrine secretory protein 55 (Nesp55), a chromogranin-like polypeptide associated with the constitutive secretory pathway but with an unknown function. Nesp is expressed in restricted brain nuclei, suggesting an involvement in specific behaviors. We have generated a knockout of Nesp55 in mice. Nesp55-deficient mice develop normally, excluding a role of this protein in the severe postnatal effects associated with imprinting of the Gnas cluster. Behavioral analysis of adult Nesp55 mutants revealed, in three separate tasks, abnormal reactivity to novel environments independent of general locomotor activity and anxiety. This phenotype may be related to prominent Nesp55 expression in the noradrenergic locus coeruleus. These results indicate a role of maternally expressed Nesp55 in controlling exploratory behavior and are the first demonstration that imprinted genes affect such a fundamental behavior.


Asunto(s)
Conducta Animal , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Impresión Genómica/genética , Locus Coeruleus/metabolismo , Asunción de Riesgos , Uracilo/análogos & derivados , Adaptación Psicológica , Animales , Química Encefálica , Cromograninas , Dopamina/análisis , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/análisis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Locus Coeruleus/química , Masculino , Ratones , Ratones Mutantes , Norepinefrina/análisis , Uracilo/análisis
14.
Arch Oral Biol ; 86: 13-17, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29132068

RESUMEN

OBJECTIVE: To explore whether there are differences in the concentration of the secretogranin II-derived peptide secretoneurin and the chromogranin B-derived peptide PE-11 between the healthy and inflamed human dental pulps. Furthermore, colocalization studies with calcitonin gene-related peptide were performed to confirm the sensory origin of the peptidergic nerves in the dental pulp. DESIGN: The concentrations of secretoneurin and PE-11 were determined by highly sensitive radioimmunoassays in extracts of dental pulps, the molecular form of secretoneurin immunoreactivities by RP-HPLC with subsequent radioimmunoassay and colocalization studies with calcitonin gene-related peptide were performed by double immunofluorescence. RESULTS: Only secretoneurin but not PE-11 was detectable by radioimmunoassays whereas nerve fibers could be made visible for both secretoneurin and PE-11. Furthermore, there was a full colocalization of secretoneurin and PE-11 with calcitonin gene-related peptide in immunohistochemical experiments. There were no differences in the concentration of secretoneurin between the healthy and inflamed human dental pulp and moreover, the characterization of the secretoneurin immunoreactivities revealed that only authentic secretoneurin was detected with the secretoneurin antibody. CONCLUSIONS: There is unequivocal evidence that secretoneurin and PE-11 are constituents of the sensory innervation of the human dental pulp and although not exclusively but are yet present in unmyelinated C-fibers which transmit predominantly nociceptive impulses. Secretoneurin might be involved in local effector functions as well, particularly in neurogenic inflammation, given that this is the case despite of unaltered levels in inflamed tissue.


Asunto(s)
Cromogranina B/inmunología , Pulpa Dental/inmunología , Neuropéptidos/inmunología , Fragmentos de Péptidos/inmunología , Pulpitis/inmunología , Secretogranina II/inmunología , Austria , Péptido Relacionado con Gen de Calcitonina/inmunología , Estudios de Casos y Controles , Cromatografía Líquida de Alta Presión , Pulpa Dental/inervación , Técnica del Anticuerpo Fluorescente , Humanos , Radioinmunoensayo
15.
Genetics ; 209(1): 233-239, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29563147

RESUMEN

Imprinted genes are expressed from one parental allele only as a consequence of epigenetic events that take place in the mammalian germ line and are thought to have evolved through intragenomic conflict between parental alleles. We demonstrate, for the first time, oppositional effects of imprinted genes on brain and behavior. Specifically, we show that mice lacking paternal Grb10 make fewer impulsive choices, with no dissociable effects on a separate measure of impulsive action. Taken together with previous work showing that mice lacking maternal Nesp55 make more impulsive choices, this suggests that impulsive choice behavior is a substrate for the action of genomic imprinting. Moreover, the contrasting effect of these two genes suggests that impulsive choices are subject to intragenomic conflict and that maternal and paternal interests pull this behavior in opposite directions. Finally, these data may also indicate that an imbalance in expression of imprinted genes contributes to pathological conditions such as gambling and drug addiction, where impulsive behavior becomes maladaptive.


Asunto(s)
Conducta Animal , Proteína Adaptadora GRB10/genética , Expresión Génica , Impresión Genómica , Análisis de Varianza , Animales , Técnica del Anticuerpo Fluorescente , Proteína Adaptadora GRB10/metabolismo , Inmunohistoquímica , Conducta Impulsiva , Masculino , Ratones
16.
Peptides ; 27(12): 3370-6, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16956695

RESUMEN

Very recently, the authors found levels of neurokinin (NK) A-like immunoreactivities in the human retina which were more than five times higher than those of substance P (SP). The present study aimed to find out how many of these immunoreactivities can be attributed to NKA and NKB and then the exact distribution pattern of both NKA and NKB was evaluated in the human retina and compared with that of SP. For this purpose, NKA-like immunoreactivities were characterized in the human retina by reversed phase HPLC followed by radioimmunoassay using the K12 antibody which recognizes both NKA and NKB. Furthermore, the retinae from both a 22- and 70-year-old donor were processed for double-immunofluorescence NKA/SP and NKB/SP. The results showed that NKA contributes to approximately two thirds and NKB to approximately one third of the immunoreactivities measured with the K12 antibody. NKA was found to be localized in sparse amacrine cells in the proximal inner nuclear layer, in displaced amacrine cells in the ganglion cell layer with processes ramifying in stratum 3 of the inner plexiform layer and also in sparse ganglion cells. By contrast, staining for NKB was only observed in ganglion cells and in the nerve fiber layer. Double-immunofluorescence revealed cellular colocalization of NKA with SP and also of NKB with SP. Thus, the levels of NKA and NKB are more than three and two times higher than those of SP, respectively. Whereas the distribution pattern of NKA is typical for neuropeptides, the localization of NKB exclusively in ganglion cells is atypical and unique.


Asunto(s)
Neuroquinina A/metabolismo , Neuroquinina B/metabolismo , Retina/metabolismo , Cromatografía Líquida de Alta Presión , Técnica del Anticuerpo Fluorescente , Humanos , Radioinmunoensayo , Sustancia P/metabolismo
17.
Neonatology ; 110(1): 14-20, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26968040

RESUMEN

BACKGROUND: Hypoxic-ischaemic encephalopathy is a major cause of neurologic impairment and mortality in neonates. Early knowledge of brain injury is important to guide therapeutic decisions and reliably inform the parents. Increased secretoneurin levels have been detected in adult patients suffering from brain injury and it has also been shown to be a promising early serum biomarker of unfavourable neurological outcome. However, no data are available in neonates. OBJECTIVE: The aim of this study was to obtain reference values for secretoneurin in healthy term neonates and then to assess the potential of this neuropeptide as a biomarker in the context of hypoxic-ischaemic encephalopathy in asphyxiated term neonates. METHODS: A total number of 139 term neonates, of which 7 were asphyxiated and 132 were healthy, were prospectively enrolled. Secretoneurin serum concentrations were assessed by radioimmunoassay. RESULTS: In healthy controls, secretoneurin serum concentrations were influenced by the mode of delivery (highest in infants born per vacuum extraction and lowest in infants born per caesarean section) and abnormal cardiotocography. In asphyxiated term neonates, secretoneurin concentrations were higher in umbilical cord blood and significantly lower 48 h after birth in comparison to healthy controls. CONCLUSION: Secretoneurin levels are elevated in cord blood in infants suffering from hypoxic-ischaemic encephalopathy following perinatal asphyxia. The potential of secretoneurin as a marker of neonatal hypoxic-ischaemic brain injury should be further evaluated in larger trials.


Asunto(s)
Asfixia Neonatal/sangre , Asfixia Neonatal/complicaciones , Lesiones Encefálicas/sangre , Hipoxia-Isquemia Encefálica/sangre , Neuropéptidos/sangre , Secretogranina II/sangre , Austria , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Sangre Fetal/química , Humanos , Lactante , Recién Nacido , Modelos Logísticos , Masculino , Embarazo , Estudios Prospectivos , Valores de Referencia , Nacimiento a Término
18.
Circulation ; 109(6): 777-83, 2004 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-14970115

RESUMEN

BACKGROUND: Secretoneurin is an abundant neuropeptide of the central, peripheral, and autonomic nervous systems, located in nerve fibers characterized by a close interaction with blood vessels and known to stimulate endothelial cell migration. METHODS AND RESULTS: We hypothesized that secretoneurin might act as an angiogenic cytokine and tested for these effects in vivo using a mouse cornea neovascularization model and in vitro by assessing capillary tube formation in a matrigel assay. In vivo, secretoneurin-induced neovasculature is characterized by a distinct pattern of arterial and venous vessels of large diameter and length. Immunohistochemical staining for CD-31 revealed endothelial lining of the inner surface of these vessels, and recruitment of alpha-smooth muscle actin-positive perivascular cells suggests vessel maturation. In vitro, secretoneurin-induced capillary tube formation was dose dependent and specific, confirming that effects of secretoneurin occur directly on endothelial cells. Secretoneurin also stimulated proliferation and exerted antiapoptotic effects on endothelial cells and activated intracellular phosphatidylinositol 3' kinase/Akt and mitogen-activated protein kinase pathways, as demonstrated by increased phosphorylation of Akt and extracellular signal-regulated kinase. CONCLUSIONS: These data show that secretoneurin represents a novel direct angiogenic cytokine and reiterate the coordinated relationship between nervous and vascular systems.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Neuropéptidos/farmacología , Proteínas Serina-Treonina Quinasas , Animales , Apoptosis/efectos de los fármacos , División Celular , Células Cultivadas , Córnea/anatomía & histología , Córnea/irrigación sanguínea , Citocinas/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Secretogranina II , Transducción de Señal
19.
Circulation ; 110(9): 1121-7, 2004 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-15326074

RESUMEN

BACKGROUND: Induction of postnatal vasculogenesis, the mobilization of bone marrow-derived endothelial progenitor cells and incorporation of these cells into sites of blood vessel formation, is a well-known feature of angiogenic cytokines such as vascular endothelial growth factor. We hypothesized that the angiogenic neuropeptide secretoneurin induces this kind of neovascularization. METHODS AND RESULTS: Secretoneurin induced mobilization of endothelial progenitor cells to sites of vasculogenesis in vivo in the cornea neovascularization assay. Progenitor cells were incorporated into vascular structures or were located adjacent to them. Systemic injection of secretoneurin led to increase of circulating stem cells and endothelial progenitor cells. In vitro secretoneurin induced migration, exerted antiapoptotic effects, and increased the number of these cells. Furthermore, secretoneurin stimulated the mitogen-activated protein kinase system, as shown by phosphorylation of extracellular signal-regulated kinase, and activated the protein kinase B/Akt pathway. Activation of mitogen-activated protein kinase was necessary for increase of cell number and migration, whereas Akt seemed to play a role in migration of endothelial progenitor cells. CONCLUSIONS: These data show that the angiogenic neuropeptide secretoneurin stimulates postnatal vasculogenesis by mobilization, migration, and incorporation of endothelial progenitor cells.


Asunto(s)
Neovascularización de la Córnea , Neuropéptidos/fisiología , Androstadienos/farmacología , Animales , Apoptosis/efectos de los fármacos , Trasplante de Médula Ósea , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Neovascularización de la Córnea/tratamiento farmacológico , Flavonoides/farmacología , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Operón Lac , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos , Ratones Transgénicos , Neuropéptidos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Quimera por Radiación , Ratas , Receptor TIE-2/genética , Secretogranina II , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Wortmanina
20.
Curr Protein Pept Sci ; 6(4): 373-85, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16101435

RESUMEN

Secretoneurin (SN) represents a 33 amino acid neuropeptide, which is highly conserved between mammals, reptiles, birds, amphibians and fish. It is specifically expressed in endocrine, neuroendocrine and neuronal tissues. In brain, the pattern of SN expression is widespread and unique, partially overlapping with established neurotransmitters. ProSN, the precursor protein, also named secretogranin II, belongs to a class of proteins collectively called chromogranins. Changes in ProSN mRNA, which is significantly regulated by cell depolarisation, represent a useful marker for both rapid and long-lasting changes (positive and negative) of neuronal activity. Under pathophysiological conditions, especially following cellular hypoxia, SN expression can be induced in non-endocrine tissues like muscle cells, pneumocytes or tumor epithelial cells. Several biological effects were attributed to SN. SN releases dopamine from rat striatal slices in a dose dependent manner and influences neurite outgrowth in the developing cerebellum. It potently and specifically attracts monocytes, eosinophils and endothelial cells towards a concentration gradient and acts as an angiogenic cytokine comparable in potency to VEGF. Thus, SN contributes to neurogenic inflammation and might play a role in the (hypoxia-driven) induction of neo-vascularisation in ischemic diseases like peripheral or coronary artery disease, diabetic retinopathia, cerebral ischemia or in solid tumors. The signalling pathways of various biological effects have not been identified in detail, but most data point to a G-protein coupled receptor structure with respective associated intracellular events.


Asunto(s)
Vasos Sanguíneos/metabolismo , Quimiotaxis/fisiología , Sistema Inmunológico/metabolismo , Neovascularización Fisiológica/fisiología , Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Animales , Vasos Sanguíneos/citología , Humanos , Sistema Inmunológico/citología , Tejido Nervioso/citología , Neuropéptidos/química , Neuropéptidos/genética , Secretogranina II , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA