Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroendocrinol ; 31(4): e12703, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30803087

RESUMEN

Angiotensin II (Ang II) acts on Ang II type 1 (AT1) receptors located in the organum vasculosum and subfornical organ (SFO) of the lamina terminalis as a main facilitatory mechanism of sodium appetite. The brain serotonin (5-HT) system with soma located in the dorsal raphe nucleus (DRN) provides a main inhibitory mechanism. In the present study, we first investigated the existence of Ang II AT1 receptors in serotonergic DRN neurones. Then, we examined whether whole body sodium depletion affects the gene expression of the AT1a receptor subtype and the presumed functional significance of AT1 receptors. Using confocal microscopy, we found that tryptophan hydroxylase-2 and serotonin neurones express AT1 receptors in the DRN. Immunofluorescence quantification showed a significant reduction in 5-HT content but no change in AT1 receptor expression or AT1/5-HT colocalisation in the DRN after sodium depletion. Whole body sodium depletion also significantly increased Agtr1a mRNA expression in the SFO and DRN. Oral treatment with the AT1 receptor antagonist losartan reversed the changes in Agtr1a expression in the SFO but not the DRN. Losartan injection into either the DRN or the mesencephalic aqueduct had no influence on sodium depletion-induced 0.3 mol L-1 NaCl intake. The results indicate the expression of Agtr1a mRNA in the DRN and SFO as a marker of sodium depletion. They also suggest that serotonergic DRN neurones are targets for Ang II. However, the function of their AT1 receptors remains elusive.


Asunto(s)
Núcleo Dorsal del Rafe/metabolismo , Expresión Génica , Receptor de Angiotensina Tipo 1/genética , Serotonina/análisis , Sodio/deficiencia , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Apetito/fisiología , Núcleo Dorsal del Rafe/química , Técnica del Anticuerpo Fluorescente , Expresión Génica/fisiología , Losartán/farmacología , Masculino , Neuronas/química , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/análisis , Receptor de Angiotensina Tipo 1/fisiología , Sodio/sangre , Órgano Subfornical/química , Órgano Subfornical/metabolismo , Triptófano Hidroxilasa/análisis
2.
Can J Physiol Pharmacol ; 96(5): 527-534, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29505736

RESUMEN

Transgenic mice constitutively overexpressing the cytokine transforming growth factor-ß1 (TGF-ß1) (TGF mice) display cerebrovascular alterations as seen in Alzheimer's disease (AD) and vascular cognitive impairment and dementia (VCID), but no or only subtle cognitive deficits. TGF-ß1 may exert part of its deleterious effects through interactions with angiotensin II (AngII) type 1 receptor (AT1R) signaling pathways. We test such interactions in the brain and cerebral vessels of TGF mice by measuring cerebrovascular reactivity, levels of protein markers of vascular fibrosis, nitric oxide synthase activity, astrogliosis, and mnemonic performance in mice treated (6 months) with the AT1R blocker losartan (10 mg/kg per day) or the angiotensin converting enzyme inhibitor enalapril (3 mg/kg per day). Both treatments restored the severely impaired cerebrovascular reactivity to acetylcholine, calcitonin gene-related peptide, endothelin-1, and the baseline availability of nitric oxide in aged TGF mice. Losartan, but not enalapril, significantly reduced astrogliosis and cerebrovascular levels of profibrotic protein connective tissue growth factor while raising levels of antifibrotic enzyme matrix metallopeptidase-9. Memory was unaffected by aging and treatments. The results suggest a pivotal role for AngII in TGF-ß1-induced cerebrovascular dysfunction and neuroinflammation through AT1R-mediated mechanisms. Further, they suggest that AngII blockers could be appropriate against vasculopathies and astrogliosis associated with AD and VCID.


Asunto(s)
Encéfalo/irrigación sanguínea , Gliosis/patología , Gliosis/fisiopatología , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Encéfalo/efectos de los fármacos , Enalapril/farmacología , Enalapril/uso terapéutico , Femenino , Fibrosis , Gliosis/metabolismo , Losartán/farmacología , Losartán/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos
3.
Am J Hypertens ; 31(2): 188-196, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28985343

RESUMEN

BACKGROUND: The right atrium is densely innervated and provides sensory input to important cardiocirculatory reflexes controlling cardiac output and blood pressure. Its angiotensin (Ang) II-expressing innervation may release Ang II as a neuropeptide cotransmitter to modulate reflexes but has not yet been characterized. METHODS: Intraoperative surgical biopsies from human right atria (n = 7) were immunocytologically stained for Ang II, tyrosine hydroxylase (TH), and synaptophysin (SYN). Tissue angiotensins were extracted and quantified by radioimmunoassay. RESULTS: Angiotensinergic fibers were frequent in epicardial nerves and around vessels with variable TH co-localization (none to >50%/bundle). Fibers were also widely distributed between cardiomyocytes and in the endocardium where they were typically nonvaricose, TH/SYN-negative and usually accompanied by varicose catecholaminergic fibers. In the endocardium, some showed large varicosities and were partially TH or SYN-positive. A few endocardial regions showed scattered nonvaricose Ang fibers ending directly between endothelial cells. Occasional clusters of thin varicose terminals co-localizing SYN or TH were located underneath, or protruded into, the endothelium. Endocardial density of Ang and TH-positive fibers was 30-300 vs. 200-450/mm2. Atrial Ang II, III, and I concentrations were 67, 16, and 5 fmol/g (median) while Ang IV and V were mostly undetectable. CONCLUSIONS: The human right atrium harbors an abundant angiotensinergic innervation and a novel potential source of atrial Ang II. Most peripheral fibers were noncatecholaminergic afferents or preterminal vagal efferents and a minority was presumably sympathetic. Neuronal Ang II release from these fibers may modulate cardiac and circulatory reflexes independently from plasma and tissue Ang II sources.


Asunto(s)
Angiotensina II/análisis , Sistema Nervioso Autónomo/química , Atrios Cardíacos/inervación , Fibras Nerviosas/química , Reflejo , Anciano , Angiotensina I/análisis , Angiotensina II/análogos & derivados , Angiotensina III/análisis , Angiotensinas/análisis , Humanos , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/análisis , Sinaptofisina/análisis , Tirosina 3-Monooxigenasa/análisis
4.
Histochem Cell Biol ; 147(5): 585-593, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28220244

RESUMEN

The renal tissular renin-angiotensin and bradykinin-kallikrein systems control kidney function together with the renal sympathetic innervation but their interaction is still unclear. To further elucidate this relationship, we investigated these systems in rats 6 days after left kidney denervation (DNX, n = 8) compared to sham-operated controls (CTR, n = 8). Plasma renin concentration was unchanged in DNX vs. CTR (p = NS). Kidney bradykinin (BK) and angiotensin (Ang) I and II concentrations decreased bilaterally in DNX vs. CTR rats (~20 to 40%, p < 0.05) together with Ang IV and V concentrations that were extremely low (p = NS). Renin, Ang III and dopamine concentrations decreased by ~25 to 50% and norepinephrine concentrations by 99% in DNX kidneys (p < 0.05) but were unaltered in opposite kidneys. Ang II/I and KA were comparable in DNX, contralateral and CTR kidneys. Ang III/II increased in right vs. DNX or CTR kidneys (40-50%, p < 0.05). Ang II was mainly located in tubular epithelium by immunocytological staining and its cellular distribution was unaffected by DNX. Moreover, the angiotensinergic and catecholaminergic innervation of right kidneys was unchanged vs. CTR. We found an important dependency of tissular Ang and BK levels on the renal innervation that may contribute to the resetting of kidney function after DNX. The DNX-induced peptide changes were not readily explained by kidney KA, renin or plasma Ang I generation. However, tissular peptide metabolism and compartmentalization may have played a central role. The mechanisms behind the concentration changes remain unclear and deserve further clarification.


Asunto(s)
Angiotensinas/metabolismo , Bradiquinina/metabolismo , Desnervación , Calicreínas/metabolismo , Riñón/inervación , Riñón/metabolismo , Renina/metabolismo , Animales , Riñón/cirugía , Masculino , Ratas , Ratas Wistar
5.
Sci Rep ; 7: 39988, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28051180

RESUMEN

Cardiac tissue engineering using biomaterials with or without combination of stem cell therapy offers a new option for repairing infarcted heart. However, the bioactivity of biomaterials remains to be optimized because currently available biomaterials do not mimic the biochemical components as well as the structural properties of native myocardial extracellular matrix. Here we hypothesized that human heart valve-derived scaffold (hHVS), as a clinically relevant novel biomaterial, may provide the proper microenvironment of native myocardial extracellular matrix for cardiac repair. In this study, human heart valve tissue was sliced into 100 µm tissue sheet by frozen-sectioning and then decellularized to form the hHVS. Upon anchoring onto the hHVS, post-infarct murine BM c-kit+ cells exhibited an increased capacity for proliferation and cardiomyogenic differentiation in vitro. When used to patch infarcted heart in a murine model of myocardial infarction, either implantation of the hHVS alone or c-kit+ cell-seeded hHVS significantly improved cardiac function and reduced infarct size; while c-kit+ cell-seeded hHVS was even superior to the hHVS alone. Thus, we have successfully developed a hHVS for cardiac repair. Our in vitro and in vivo observations provide the first clinically relevant evidence for translating the hHVS-based biomaterials into clinical strategies to treat myocardial infarction.


Asunto(s)
Células de la Médula Ósea/fisiología , Válvula Mitral/fisiología , Infarto del Miocardio/terapia , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Materiales Biocompatibles , Células de la Médula Ósea/ultraestructura , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Válvula Mitral/ultraestructura
6.
J Cereb Blood Flow Metab ; 37(6): 1959-1970, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27389178

RESUMEN

Alterations of the renin-angiotensin system have been implicated in the pathogenesis of Alzheimer's disease. We tested the efficacy of losartan (10 mg/kg/day for three months), a selective angiotensin II type 1 receptor antagonist, in alleviating cerebrovascular and cognitive deficits in double-transgenic mice (six months at endpoint) that overexpress a mutated form of the human amyloid precursor protein (APPSwe,Ind) and a constitutively active form of the transforming growth factor-ß1, thereafter named A/T mice. Losartan rescued cerebrovascular reactivity, particularly the dilatory responses, but failed to attenuate astroglial activation and to normalize the neurovascular uncoupling response to sensory stimulation. The cognitive deficits of A/T mice were not restored by losartan nor were the increased brain levels of soluble and insoluble Aß1-40 and Aß1-42 peptides normalized. Our results are the first to demonstrate the capacity of losartan to improve cerebrovascular reactivity in an Alzheimer's disease mouse model of combined Aß-induced vascular oxidative stress and transforming growth factor-ß1-mediated vascular fibrosis. These data suggest that losartan may be promising for restoring cerebrovascular function in patients with vascular diseases at risk for vascular dementia or Alzheimer's disease. However, a combined therapy may be warranted for rescuing both vascular and cognitive deficits in a multifaceted pathology like Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/biosíntesis , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Circulación Cerebrovascular/efectos de los fármacos , Losartán/uso terapéutico , Factor de Crecimiento Transformador beta1/biosíntesis , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Animales , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Humanos , Losartán/administración & dosificación , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Estrés Oxidativo/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Factor de Crecimiento Transformador beta1/genética
7.
J Alzheimers Dis ; 51(4): 1183-95, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26923013

RESUMEN

The co-administration of angiotensin converting enzyme inhibitors (ACEi) and angiotensin II (AngII) receptor blockers (ARB) that bind angiotensin type 1 receptors (AT1R) may protect from Alzheimer's disease (AD) better than each treatment taken alone. We tested the curative potential of the non brain-penetrant ACEi enalapril (3 mg/kg/day) administered for 3 months either alone or in combination with the brain penetrant ARB losartan (10 mg/kg/day) in aged (∼15 months) transgenic mice overexpressing a mutated form of the human amyloid-ß protein precursor (AßPP, thereafter APP mice). We studied cerebrovascular function, protein levels of oxidative stress markers (superoxide dismutases SOD1, SOD2 and the NADPH oxidase subunit p67phox), amyloid-ß (Aß) pathology, astrogliosis, cholinergic innervation, AT1R and angiotensin IV receptor (AT4R) levels, together with cognitive performance. Both treatments normalized cerebrovascular reactivity and p67phox protein levels, but they did not reduce the cerebrovascular levels of SOD1. Combined treatment normalized cerebrovascular SOD2 levels, significantly attenuated astrogliosis, but did not reduce the increased levels of cerebrovascular AT1R. Yet, combined therapy enhanced thioflavin-S labeled Aß plaque burden, a tendency not significant when Aß1 - 42 plaque load was considered. None of the treatments rescued cognitive deficits, cortical AT4R or cholinergic innervation. We conclude that both treatments normalized cerebrovascular function by inhibiting the AngII-induced oxidative stress cascade, and that the positive effects of the combined therapy on astrogliosis were likely due to the ability of losartan to enter brain parenchyma. However, enalapril did not potentiate, and may even dampen, the reported cognitive benefits of losartan, raising caution when selecting the most appropriate antihypertensive therapy in AD patients.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Antihipertensivos/uso terapéutico , Trastornos Cerebrovasculares/tratamiento farmacológico , Trastornos Cerebrovasculares/etiología , Enalapril/uso terapéutico , Losartán/uso terapéutico , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/tratamiento farmacológico , Amiloidosis/etiología , Análisis de Varianza , Animales , Colinesterasas/metabolismo , Modelos Animales de Enfermedad , Combinación de Medicamentos , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Ratones , Ratones Transgénicos , Mutación/genética
8.
Behav Brain Res ; 272: 314-23, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25046593

RESUMEN

A single or repeated exposure to psychostimulants induces long-lasting neuroadaptative changes. Different neurotransmitter systems are involved in these responses including the neuropeptide angiotensin II. Our study tested the hypothesis that the neuroadaptative changes induced by amphetamine produce alterations in brain RAS components that are involved in the expression of the locomotor sensitization to the psychostimulant drug. Wistar male rats, pretreated with amphetamine were used 7 or 21 days later to study AT1 receptors by immunohistochemistry and western blot and also angiotensinogen mRNA and protein in caudate putamen and nucleus accumbens. A second group of animals was used to explore the possible role of Ang II AT1 receptors in the expression of behavioral sensitization. In these animals treated in the same way, bearing intra-cerebral cannula, the locomotor activity was tested 21 days later, after an amphetamine challenge injection and the animals received an AT1 blocker, losartan, or saline 5min before the amphetamine challenge. An increase of AT1 receptor density induced by amphetamine was found in both studied areas and a decrease in angiotensinogen mRNA and protein only in CPu at 21 days after treatment; meanwhile, no changes were established in NAcc. Finally, the increased locomotor activity induced by amphetamine challenge was blunted by losartan administration in CPu. No differences were detected in the behavioral sensitization when the AT1 blocker was injected in NAcc. Our results support the hypothesis of a key role of brain RAS in the neuroadaptative changes induced by amphetamine.


Asunto(s)
Anfetamina/farmacología , Encéfalo/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Angiotensinógeno/metabolismo , Animales , Encéfalo/fisiología , Losartán/farmacología , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas Wistar , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina/fisiología
9.
Neurobiol Dis ; 68: 126-36, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24807206

RESUMEN

Angiotensin II (AngII) receptor blockers that bind selectively AngII type 1 (AT1) receptors may protect from Alzheimer's disease (AD). We studied the ability of the AT1 receptor antagonist losartan to cure or prevent AD hallmarks in aged (~18months at endpoint, 3months treatment) or adult (~12months at endpoint, 10months treatment) human amyloid precursor protein (APP) transgenic mice. We tested learning and memory with the Morris water maze, and evaluated neurometabolic and neurovascular coupling using [(18)F]fluoro-2-deoxy-D-glucose-PET and laser Doppler flowmetry responses to whisker stimulation. Cerebrovascular reactivity was assessed with on-line videomicroscopy. We measured protein levels of oxidative stress enzymes (superoxide dismutases SOD1, SOD2 and NADPH oxidase subunit p67phox), and quantified soluble and deposited amyloid-ß (Aß) peptide, glial fibrillary acidic protein (GFAP), AngII receptors AT1 and AT2, angiotensin IV receptor AT4, and cortical cholinergic innervation. In aged APP mice, losartan did not improve learning but it consolidated memory acquisition and recall, and rescued neurovascular and neurometabolic coupling and cerebrovascular dilatory capacity. Losartan normalized cerebrovascular p67phox and SOD2 protein levels and up-regulated those of SOD1. Losartan attenuated astrogliosis, normalized AT1 and AT4 receptor levels, but failed to rescue the cholinergic deficit and the Aß pathology. Given preventively, losartan protected cognitive function, cerebrovascular reactivity, and AT4 receptor levels. Like in aged APP mice, these benefits occurred without a decrease in soluble Aß species or plaque load. We conclude that losartan exerts potent preventive and restorative effects on AD hallmarks, possibly by mitigating AT1-initiated oxidative stress and normalizing memory-related AT4 receptors.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Circulación Cerebrovascular/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Losartán/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Arginina/análogos & derivados , Arginina/farmacología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Endotelina-1/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Losartán/farmacología , Masculino , Ratones , Ratones Transgénicos , Mutación/genética
10.
Curr Hypertens Rep ; 15(1): 10-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23197298

RESUMEN

Intrarenal neurotransmission implies the co-release of neuropeptides at the neuro-effector junction with direct influence on parameters of kidney function. The presence of an angiotensin (Ang) II-containing phenotype in catecholaminergic postganglionic and sensory fibers of the kidney, based on immunocytological investigations, has only recently been reported. These angiotensinergic fibers display a distinct morphology and intrarenal distribution, suggesting anatomical and functional subspecialization linked to neuronal Ang II-expression. This review discusses the present knowledge concerning these fibers, and their significance for renal physiology and the pathogenesis of hypertension in light of established mechanisms. The data suggest a new role of Ang II as a co-transmitter stimulating renal target cells or modulating nerve traffic from or to the kidney. Neuronal Ang II is likely to be an independent source of intrarenal Ang II. Further physiological experimentation will have to explore the role of the angiotensinergic renal innervation and integrate it into existing concepts.


Asunto(s)
Angiotensina II/fisiología , Riñón/inervación , Riñón/fisiología , Animales , Sistema Nervioso Autónomo/fisiología , Endotelio Vascular/fisiología , Humanos , Hipertensión/metabolismo , Inmunohistoquímica , Riñón/metabolismo , Unión Neuroefectora/fisiología , Neuronas/química , Neuropéptidos/fisiología , Transmisión Sináptica/fisiología
11.
Histol Histopathol ; 27(11): 1413-28, 2012 11.
Artículo en Inglés | MEDLINE | ID: mdl-23018241

RESUMEN

We describe an angiotensin (Ang) II-containing innervation of the kidney. Cryosections of rat, pig and human kidneys were investigated for the presence of Ang II-containing nerve fibers using a mouse monoclonal antibody against Ang II (4B3). Co-staining was performed with antibodies against synaptophysin, tyrosine 3-hydroxylase, and dopamine beta-hydroxylase to detect catecholaminergic efferent fibers and against calcitonin gene-related peptide to detect sensory fibers. Tagged secondary antibodies and confocal light or laser scanning microscopy were used for immunofluorescence detection. Ang II-containing nerve fibers were densely present in the renal pelvis, the subepithelial layer of the urothelium, the arterial nervous plexus, and the peritubular interstitium of the cortex and outer medulla. They were infrequent in central veins and the renal capsule and absent within glomeruli and the renal papilla. Ang II-positive fibers represented phenotypic subgroups of catecholaminergic postganglionic or sensory fibers with different morphology and intrarenal distribution compared to their Ang II-negative counterparts. The Ang II-positive postganglionic fibers were thicker, produced typically fusiform varicosities and preferentially innervated the outer medulla and periglomerular arterioles. Ang II-negative sensory fibers were highly varicose, prevailing in the pelvis and scarce in the renal periphery compared to the rarely varicose Ang II-positive fibers. Neurons within renal microganglia displayed angiotensinergic, catecholaminergic, or combined phenotypes. Our results suggest that autonomic fibers may be an independent source of intrarenal Ang II acting as a neuropeptide co-transmitter or neuromodulator. The angiotensinergic renal innervation may play a distinct role in the neuronal control of renal sodium reabsorption, vasomotion and renin secretion.


Asunto(s)
Angiotensina II/metabolismo , Riñón/inervación , Fibras Nerviosas/metabolismo , Neuronas/metabolismo , Animales , Sistema Nervioso Autónomo/metabolismo , Humanos , Riñón/metabolismo , Masculino , Ratas , Ratas Endogámicas WKY , Porcinos , Urotelio/metabolismo
12.
Front Biosci (Landmark Ed) ; 17(7): 2419-32, 2012 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-22652789

RESUMEN

Angiotensin (Ang) II has for long been identified as a neuropeptide located within neurons and pathways of the central nervous system involved in the control of thirst and cardio-vascular homeostasis. The presence of Ang II in ganglionic neurons of celiac, dorsal root, and trigeminal ganglia has only recently been described in humans and rats. Ang II-containing fibers were also found in the mesenteric artery and the heart, together with intrinsic Ang II-containing cardiac neurons. Ganglionic neurons express angiotensinogen and co-localize it with Ang II. Its intraneuronal production as a neuropeptide appears to involve angiotensinogen processing enzymes other than renin. Immunocytochemical and gene expression data suggest that neuronal Ang II acts as a neuromodulatory peptide and co-transmitter in the peripheral autonomic, and also sensory nervous system. Neuronal Ang II probably competes with humoral Ang II for effector cell activation. Its functional role, however, still remains to be determined. Angiotensinergic neurotransmission in the autonomic nervous system is a potential new target for therapeutic interventions in many common diseases such as essential hypertension, heart failure, and cardiac arrhythmia.


Asunto(s)
Angiotensina II/fisiología , Sistema Nervioso Autónomo/fisiología , Transmisión Sináptica/fisiología , Animales , Encéfalo/fisiología , Ganglios Espinales/fisiología , Corazón/inervación , Humanos , Modelos Neurológicos , Neuropéptidos/fisiología , Ratas , Renina/fisiología , Células Receptoras Sensoriales/fisiología , Sistema Nervioso Simpático/fisiología , Ganglio del Trigémino/fisiología
13.
Regul Pept ; 167(1): 31-41, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21145919

RESUMEN

Although the physiological and pharmacological evidences suggest a role for angiotensin II (Ang II) with the mammalian heart, the source and precise location of Ang II are unknown. To visualize and quantitate Ang II in atria, ventricular walls and interventricular septum of the rat and human heart and to explore the feasibility of local Ang II production and function, we investigated by different methods the expression of proteins involved in the generation and function of Ang II. We found mRNA of angiotensinogen (Ang-N), of angiotensin converting enzyme, of the angiotensin type receptors AT(1A) and AT2 (AT(1B) not detected) as well as of cathepsin D in any part of the hearts. No renin mRNA was traceable. Ang-N mRNA was visualized by in situ hybridization in atrial ganglial neurons. Ang II and dopamine-ß-hydroxylase (DßH) were either colocalized inside the same neuronal cell or the neurons were specialized for Ang II or DßH. Within these neurons, the vesicular acetylcholine transporter (VAChT) was neither colocalized with Ang II nor DßH, but VAChT-staining was found with synapses en passant encircle these neuronal cells. The fibers containing Ang II exhibited with blood vessels and with cardiomyocytes supposedly angiotensinergic synapses en passant. In rat heart, right atrial median Ang II concentration appeared higher than septal and ventricular Ang II. The distinct colocalization of neuronal Ang II with DßH in the heart may indicate that Ang II participates together with norepinephrine in the regulation of cardiac functions: produced as a cardiac neurotransmitter Ang II may have inotropic, chronotropic or dromotropic effects in atria and ventricles and contributes to blood pressure regulation.


Asunto(s)
Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Dopamina beta-Hidroxilasa/metabolismo , Neuronas/fisiología , Neurotransmisores/metabolismo , Sinapsis/fisiología , Anciano , Anciano de 80 o más Años , Angiotensina II/genética , Angiotensinógeno/genética , Animales , Presión Sanguínea/fisiología , Catepsina D/genética , Catepsina D/metabolismo , Dopamina beta-Hidroxilasa/genética , Femenino , Expresión Génica , Atrios Cardíacos/metabolismo , Atrios Cardíacos/ultraestructura , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Persona de Mediana Edad , Neuronas/ultraestructura , Neurotransmisores/genética , Norepinefrina/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Endogámicas WKY , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Sinapsis/ultraestructura , Tabique Interventricular/fisiología , Tabique Interventricular/ultraestructura , Proteínas de Transporte Vesicular de Acetilcolina/genética , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
14.
J Immunol ; 185(10): 6286-93, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20935205

RESUMEN

Emerging evidence suggests a cardioprotective role of the angiotensin AT2R, albeit the underlying cellular mechanisms are not well understood. We aimed in this article to elucidate a potential role of cardiac angiotensin AT2R in regulating cellular immune response to ischemic heart injury. Seven days after myocardial infarction in rats, double-immunofluorescence staining showed that AT2R was detected in a fraction of CD8(+) T cells infiltrating in the peri-infarct myocardium. We developed a method that allowed the isolation of myocardial infiltrating CD8(+)AT2R(+) T cells using modified MACS, and further characterization and purification with flow cytometry. Although the CD8(+)AT2R(-) T cells exhibited potent cytotoxicity to both adult and fetal cardiomyocytes (CMs), the CD8(+)AT2R(+) T cells were noncytotoxic to these CMs. The CD8(+)AT2R(+) T cells were characterized by upregulated IL-10 and downregulated IL-2 and INF-γ expression when compared with CD8(+)AT2R(-) T cells. We further showed that IL-10 gene expression was enhanced in CD8(+) T cells on in vitro AT2R stimulation. Importantly, in vivo AT2R activation engendered an increment of CD8(+)AT2R(+) T cells and IL-10 production in the ischemic myocardium. In addition, intramyocardial transplantation of CD8(+)AT2R(+) T cells (versus CD8(+)AT2R(-)) led to reduced ischemic heart injury. Moreover, the CD8(+)AT2R(+) T cell population was also demonstrated in human peripheral blood. Thus, we have defined the cardioprotective CD8(+)AT2R(+) T cell population, which increases during ischemic heart injury and contributes to maintaining CM viability and providing IL-10, hence revealing an AT2R-mediated cellular mechanism in modulating adaptive immune response in the heart.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucina-10/biosíntesis , Infarto del Miocardio/inmunología , Miocardio/inmunología , Receptor de Angiotensina Tipo 2/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Separación Celular/métodos , Citometría de Flujo/métodos , Técnica del Anticuerpo Fluorescente , Expresión Génica , Interleucina-10/inmunología , Masculino , Infarto del Miocardio/metabolismo , Isquemia Miocárdica/inmunología , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología
15.
Cell Tissue Res ; 341(1): 181-95, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20526894

RESUMEN

The angiotensins constitute a neuropeptidergic system that emerged early in evolution. Their classical osmoregulatory and dipsogenic functions and their mnemonic actions have been demonstrated both in vertebrates and in some invertebrates. Previously, we have shown that, in the euryhaline and semiterrestrial crab Chasmagnathus granulatus, water deprivation correlates with an increased level of brain angiotensin-II-like neuropeptide/s (ANGII-like) and improves memory processes through ANGII receptors. We have proposed that the release of brain angiotensins in response to water shortages is an ancient mechanism for coordinating various functions that, together, enable organisms to tolerate this environmental change. Here, we have evaluated the physiological changes in ANGII-like levels in diverse structures of the central nervous system of these animals during water deprivation. The neuroanatomical distribution of ANGII-like is described in the optic lobes and brain of Chasmagnathus granulatus and the physiological changes in ANGII-like distribution in various brain neuropils is evaluated after water deprivation. Our results indicate that ANGII-like is widely distributed, especially in the medial protocerebrum. After 2 h of water deprivation, ANGII-like immunoreactivity increases in the central body and decreases in the olfactory neuropil and, after 6 h of water deprivation, is markedly reduced in several brain areas. Although further experiments are needed to establish that the angiotensinergic system is involved in the balance of body fluids in this crab, our results suggest that ANGII regulates several functions during water shortages.


Asunto(s)
Angiotensina II/metabolismo , Braquiuros/anatomía & histología , Braquiuros/fisiología , Sistema Nervioso Central/anatomía & histología , Sistema Nervioso Central/fisiología , Privación de Agua/fisiología , Animales , Braquiuros/citología , Encéfalo/anatomía & histología , Encéfalo/fisiología , Sistema Nervioso Central/citología , Inmunohistoquímica , Masculino , Lóbulo Óptico de Animales no Mamíferos/anatomía & histología , Lóbulo Óptico de Animales no Mamíferos/fisiología , Transporte de Proteínas
16.
Regul Pept ; 162(1-3): 90-8, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20346377

RESUMEN

To elucidate the local formation of angiotensin II (Ang II) in the neurons of sensory dorsal root ganglia (DRG), we studied the expression of angiotensinogen (Ang-N)-, renin-, angiotensin converting enzyme (ACE)- and cathepsin D-mRNA, and the presence of protein renin, Ang II, Substance P and calcitonin gene-related peptide (CGRP) in the rat and human thoracic DRG. Quantitative real time PCR (qRT-PCR) studies revealed that rat DRG expressed substantial amounts of Ang-N- and ACE mRNA, while renin mRNA as well as the protein renin were untraceable. Cathepsin D-mRNA and cathepsin D-protein were detected in the rat DRG indicating the possibility of existence of pathways alternative to renin for Ang I formation. Angiotensin peptides were successfully detected with high performance liquid chromatography and radioimmunoassay in human DRG extracts. In situ hybridization in rat DRG confirmed additionally expression of Ang-N mRNA in the cytoplasm of numerous neurons. Intracellular Ang II staining could be shown in number of neurons and their processes in both the rat and human DRG. Interestingly we observed neuronal processes with angiotensinergic synapses en passant, colocalized with synaptophysin, within the DRG. In the DRG, we also identified by qRT-PCR, expression of Ang II receptor AT(1A) and AT(2)-mRNA while AT(1B)-mRNA was not traceable. In some neurons Substance P and CGRP were found colocalized with Ang II. The intracellular localization and colocalization of Ang II with Substance P and CGRP in the DRG neurons may indicate a participation and function of Ang II in the regulation of nociception. In conclusion, these results suggest that Ang II may be produced locally in the neurons of rat and human DRG and act as a neurotransmitter.


Asunto(s)
Angiotensinas/metabolismo , Ganglios Espinales/metabolismo , Neuronas/metabolismo , Angiotensinógeno/genética , Animales , Secuencia de Bases , Catepsina D/genética , Cromatografía Líquida de Alta Presión , Cartilla de ADN , Humanos , Inmunohistoquímica , Masculino , Peptidil-Dipeptidasa A/genética , ARN Mensajero/genética , Ratas , Ratas Endogámicas WKY , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Stem Cells ; 27(10): 2488-97, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19591228

RESUMEN

The expression pattern of angiotensin AT2 receptors with predominance during fetal life and upregulation under pathological conditions during tissue injury/repair process suggests that AT2 receptors may exert an important action in injury/repair adaptive mechanisms. Less is known about AT2 receptors in acute ischemia-induced cardiac injury. We aimed here to elucidate the role of AT2 receptors after acute myocardial infarction. Double immunofluorescence staining showed that cardiac AT2 receptors were mainly detected in clusters of small c-kit+ cells accumulating in peri-infarct zone and c-kit+AT2+ cells increased in response to acute cardiac injury. Further, we isolated cardiac c-kit+AT2+ cell population by modified magnetic activated cell sorting and fluorescence activated cell sorting. These cardiac c-kit+AT2+ cells, represented approximately 0.19% of total cardiac cells in infarcted heart, were characterized by upregulated transcription factors implicated in cardiogenic differentiation (Gata-4, Notch-2, Nkx-2.5) and genes required for self-renewal (Tbx-3, c-Myc, Akt). When adult cardiomyocytes and cardiac c-kit+AT2+ cells isolated from infarcted rat hearts were cocultured, AT2 receptor stimulation in vitro inhibited apoptosis of these cocultured cardiomyocytes. Moreover, in vivo AT2 receptor stimulation led to an increased c-kit+AT2+ cell population in the infarcted myocardium and reduced apoptosis of cardiomyocytes in rats with acute myocardial infarction. These data suggest that cardiac c-kit+AT2+ cell population exists and increases after acute ischemic injury. AT2 receptor activation supports performance of cardiomyocytes, thus contributing to cardioprotection via cardiac c-kit+AT2+ cell population.


Asunto(s)
Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Regeneración/fisiología , Células Madre/metabolismo , Angiotensinas/metabolismo , Angiotensinas/farmacología , Animales , Cardiotónicos/metabolismo , Cardiotónicos/farmacología , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Masculino , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/citología , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Células Madre/citología , Factores de Transcripción/metabolismo
18.
Regul Pept ; 154(1-3): 23-31, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19323983

RESUMEN

To clarify the role of Angiotensin II (Ang II) in the sensory system and especially in the trigeminal ganglia, we studied the expression of angiotensinogen (Ang-N)-, renin-, angiotensin converting enzyme (ACE)- and cathepsin D-mRNA, and the presence of Ang II and substance P in the rat and human trigeminal ganglia. The rat trigeminal ganglia expressed substantial amounts of Ang-N- and ACE mRNA as determined by quantitative real time PCR. Renin mRNA was untraceable in rat samples. Cathepsin D was detected in the rat trigeminal ganglia indicating the possibility of existence of pathways alternative to renin for Ang I formation. In situ hybridization in rat trigeminal ganglia revealed expression of Ang-N mRNA in the cytoplasm of numerous neurons. By using immunocytochemistry, a number of neurons and their processes in both the rat and human trigeminal ganglia were stained for Ang II. Post in situ hybridization immunocytochemistry reveals that in the rat trigeminal ganglia some, but not all Ang-N mRNA-positive neurons marked for Ang II. In some neurons Substance P was found colocalized with Ang II. Angiotensins from rat trigeminal ganglia were quantitated by radioimmunoassay with and without prior separation by high performance liquid chromatography. Immunoreactive angiotensin II (ir-Ang II) was consistently present and the sum of true Ang II (1-8) octapeptide and its specifically measured metabolites were found to account for it. Radioimmunological and immunocytochemical evidence of ir-Ang II in neuronal tissue is compatible with Ang II as a neurotransmitter. In conclusion, these results suggest that Ang II could be produced locally in the neurons of rat trigeminal ganglia. The localization and colocalization of neuronal Ang II with Substance P in the trigeminal ganglia neurons may be the basis for a participation and function of Ang II in the regulation of nociception and migraine pathology.


Asunto(s)
Angiotensinógeno/análisis , Angiotensinógeno/metabolismo , Neuronas/metabolismo , Ganglio del Trigémino/metabolismo , Adulto , Angiotensina I/análisis , Angiotensina II/análisis , Angiotensinógeno/genética , Animales , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , ARN Mensajero/metabolismo , Radioinmunoensayo , Ratas , Ratas Endogámicas WKY
19.
Hum Mol Genet ; 18(3): 546-55, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19010792

RESUMEN

In spinal muscular atrophy (SMA), the leading genetic cause of early childhood death, the survival motor neuron 1 gene (SMN1) is deleted or inactivated. The nearly identical SMN2 gene has a silent mutation that impairs the utilization of exon 7 and the production of functional protein. It has been hypothesized that therapies boosting SMN2 exon 7 inclusion might prevent or cure SMA. Exon 7 inclusion can be stimulated in cell culture by oligonucleotides or intracellularly expressed RNAs, but evidence for an in vivo improvement of SMA symptoms is lacking. Here, we unambiguously confirm the above hypothesis by showing that a bifunctional U7 snRNA that stimulates exon 7 inclusion, when introduced by germline transgenesis, can efficiently complement the most severe mouse SMA model. These results are significant for the development of a somatic SMA therapy, but may also provide new means to study pathophysiological aspects of this devastating disease.


Asunto(s)
Terapia Genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , ARN Nuclear Pequeño/uso terapéutico , Animales , Secuencia de Bases , Exones , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Atrofia Muscular Espinal/metabolismo , Empalme del ARN , ARN Nuclear Pequeño/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
20.
Regul Pept ; 147(1-3): 82-7, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18308407

RESUMEN

In contrast to the current belief that angiotensin II (Ang II) interacts with the sympathetic nervous system only as a circulating hormone, we document here the existence of endogenous Ang II in the neurons of rat and human sympathetic coeliac ganglia and their angiotensinergic innervation with mesenteric resistance blood vessels. Angiotensinogen - and angiotensin converting enzyme-mRNA were detected by using quantitative real time polymerase chain reaction in total RNA extracts of rat coeliac ganglia, while renin mRNA was untraceable. Cathepsin D, a protease responsible for cleavage beneath other substrates also angiotensinogen to angiotensin I, was successfully detected in rat coeliac ganglia indicating the possibility of existence of alternative pathways. Angiotensinogen mRNA was also detected by in situ hybridization in the cytoplasm of neurons of rat coeliac ganglia. Immunoreactivity for Ang II was demonstrated in rat and human coeliac ganglia as well as with mesenteric resistance blood vessels. By using confocal laser scanning microscopy we were able to demonstrate the presence of angiotensinergic synapses en passant along side of vascular smooth muscle cells. Our findings indicate that Ang II is synthesized inside the neurons of sympathetic coeliac ganglia and may act as an endogenous neurotransmitter locally with the mesenteric resistance blood vessels.


Asunto(s)
Angiotensinógeno/metabolismo , Ganglios Simpáticos/metabolismo , Arterias Mesentéricas/inervación , Neuronas/metabolismo , Angiotensinógeno/genética , Animales , Humanos , Inmunohistoquímica , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas WKY , Sistema Renina-Angiotensina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...