Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Neurosci ; 39(41): 8038-8050, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31471471

RESUMEN

Integration and modulation of primary afferent sensory information begins at the first terminating sites within the CNS, where central inhibitory circuits play an integral role. Viscerosensory information is conveyed to the nucleus of the solitary tract (NTS) where it initiates neuroendocrine, behavioral, and autonomic reflex responses that ensure optimal internal organ function. This excitatory input is modulated by diverse, local inhibitory interneurons, whose functions are not clearly understood. Here we show that, in male rats, 65% of somatostatin-expressing (SST) NTS neurons also express GAD67, supporting their likely role as inhibitory interneurons. Using whole-cell recordings of NTS neurons, from horizontal brainstem slices of male and female SST-yellow fluorescent protein (YFP) and SST-channelrhodopsin 2 (ChR2)-YFP mice, we quantified the impact of SST-NTS neurons on viscerosensory processing. Light-evoked excitatory photocurrents were reliably obtained from SST-ChR2-YFP neurons (n = 16) and the stimulation-response characteristics determined. Most SST neurons (57%) received direct input from solitary tract (ST) afferents, indicating that they form part of a feedforward circuit. All recorded SST-negative NTS neurons (n = 72) received SST-ChR2 input. ChR2-evoked PSCs were largely inhibitory and, in contrast to previous reports, were mediated by both GABA and glycine. When timed to coincide, the ChR2-activated SST input suppressed ST-evoked action potentials at second-order NTS neurons, demonstrating strong modulation of primary viscerosensory input. These data indicate that the SST inhibitory network innervates broadly within the NTS, with the potential to gate viscerosensory input to powerfully alter autonomic reflex function and other behaviors.SIGNIFICANCE STATEMENT Sensory afferent input is modulated according to state. For example the baroreflex is altered during a stress response or exercise, but the basic mechanisms underpinning this sensory modulation are not fully understood in any sensory system. Here we demonstrate that the neuronal processing of viscerosensory information begins with synaptic gating at the first central synapse with second-order neurons in the NTS. These data reveal that the somatostatin subclass of inhibitory interneurons are driven by visceral sensory input to play a major role in gating viscerosensory signals, placing them within a feedforward circuit within the NTS.


Asunto(s)
Red Nerviosa/fisiología , Neuronas/fisiología , Sensación/fisiología , Filtrado Sensorial/fisiología , Somatostatina/fisiología , Animales , Retroalimentación Fisiológica , Femenino , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/fisiología , Glicina/fisiología , Interneuronas/fisiología , Masculino , Ratones , Red Nerviosa/citología , Estimulación Luminosa , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/citología , Núcleo Solitario/fisiología , Aferentes Viscerales/fisiología , Ácido gamma-Aminobutírico/fisiología
2.
J Neurosci ; 34(45): 15110-22, 2014 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-25378174

RESUMEN

Brainstem catecholaminergic neurons play key roles in the autonomic, neuroendocrine, and behavioral responses to glucoprivation, yet the functions of the individual groups are not fully understood. Adrenergic C3 neurons project widely throughout the brain, including densely to sympathetic preganglionic neurons in the spinal cord, yet their function is completely unknown. Here we demonstrate in rats that optogenetic stimulation of C3 neurons induces sympathoexcitatory, cardiovasomotor functions. These neurons are activated by glucoprivation, but unlike the C1 cell group, not by hypotension. The cardiovascular activation induced by C3 neurons is less than that induced by optogenetic stimulation of C1 neurons; however, combined stimulation produces additive sympathoexcitatory and cardiovascular effects. The varicose axons of C3 neurons largely overlap with those of C1 neurons in the region of sympathetic preganglionic neurons in the spinal cord; however, regional differences point to effects on different sympathetic outflows. These studies definitively demonstrate the first known function of C3 neurons as unique cardiovasomotor stimulatory cells, embedded in the brainstem networks regulating cardiorespiratory activity and the response to glucoprivation.


Asunto(s)
Fibras Adrenérgicas/fisiología , Tronco Encefálico/fisiología , Glucosa/metabolismo , Corazón/inervación , Sistema Nervioso Simpático/fisiología , Potenciales de Acción , Fibras Adrenérgicas/metabolismo , Animales , Tronco Encefálico/citología , Tronco Encefálico/metabolismo , Corazón/fisiología , Homeostasis , Masculino , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/metabolismo
3.
Exp Physiol ; 99(12): 1597-610, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25239924

RESUMEN

Chronic low-dose systemic infusion of angiotensin II induces hypertension via activation of the angiotensin II type 1A receptor (AT1AR). Previously, we have demonstrated that expression of the AT1AR on catecholaminergic neurons is necessary for the full development of angiotensin-dependent hypertension. In the present study, we examined the mechanism by which selective deletion of the AT1AR from these cells affects the development of hypertension. We also tested the hypothesis that AT1ARs expressed by catecholaminergic C1 neurons in the rostral ventrolateral medulla play an important role in angiotensin-induced hypertension. A Cre-lox approach was used to delete the AT1AR from all catecholaminergic cells or from C1 neurons selectively. Subcutaneous administration of angiotensin II induced hypertension in all mice, with delayed onset and reduced maximal response in the global AT1AR catecholaminergic knockout mice. The AT1AR catecholaminergic knockout mice had decreased renal fluid and electrolyte retention and urinary noradrenaline excretion. The blood pressure response was reduced only during the second week of angiotensin II infusion in the mice with selective C1 AT1AR deletion, demonstrating that AT1AR expression by C1 neurons plays a moderate role in angiotensin-induced hypertension. The difference in the time course of development of hypertension between the mice with global AT1AR knockout from catecholaminergic cells and the mice with C1 AT1AR deletion suggests that other catecholaminergic neurons are important.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión/metabolismo , Bulbo Raquídeo/metabolismo , Neuronas/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina II , Animales , Hipertensión/inducido químicamente , Hipertensión/genética , Ratones , Ratones Noqueados , Receptor de Angiotensina Tipo 1/genética
4.
J Neurosci ; 32(6): 2051-61, 2012 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-22323719

RESUMEN

The rise in blood pressure during an acute aversive stress has been suggested to involve activation of angiotensin type 1A receptors (AT(1A)Rs) at various sites within the brain, including the rostral ventrolateral medulla. In this study we examine the involvement of AT(1A)Rs associated with a subclass of sympathetic premotor neurons of the rostral ventrolateral medulla, the C1 neurons. The distribution of putative AT(1A)R-expressing cells was mapped throughout the brains of three transgenic mice with a bacterial artificial chromosome-expressing green fluorescent protein under the control of the AT(1A)R promoter. The overall distribution correlated with that of the AT(1A)Rs mapped by other methods and demonstrated that the majority of C1 neurons express the AT(1A)R. Cre-recombinase expression in C1 neurons of AT(1A)R-floxed mice enabled demonstration that the pressor response to microinjection of angiotensin II into the rostral ventrolateral medulla is dependent upon expression of the AT(1A)R in these neurons. Lentiviral-induced expression of wild-type AT(1A)Rs in C1 neurons of global AT(1A)R knock-out mice, implanted with radiotelemeter devices for recording blood pressure, modulated the pressor response to aversive stress. During prolonged cage-switch stress, expression of AT(1A)Rs in C1 neurons induced a greater sustained pressor response when compared to the control viral-injected group (22 ± 4 mmHg for AT(1A)R vs 10 ± 1 mmHg for GFP; p < 0.001), which was restored toward that of the wild-type group (28 ± 2 mmHg). This study demonstrates that AT(1A)R expression by C1 neurons is essential for the pressor response to angiotensin II and that this pathway plays an important role in the pressor response to aversive stress.


Asunto(s)
Angiotensina II/fisiología , Bulbo Raquídeo/metabolismo , Neuronas Motoras/fisiología , Presorreceptores/fisiología , Receptor de Angiotensina Tipo 1/biosíntesis , Estrés Psicológico/metabolismo , Animales , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas Motoras/patología , Receptor de Angiotensina Tipo 1/agonistas , Estrés Psicológico/patología , Estrés Psicológico/psicología
5.
Elife ; 122023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37772793

RESUMEN

The pre-Bötzinger complex (preBötC), a key primary generator of the inspiratory breathing rhythm, contains neurons that project directly to facial nucleus (7n) motoneurons to coordinate orofacial and nasofacial activity. To further understand the identity of 7n-projecting preBötC neurons, we used a combination of optogenetic viral transgenic approaches to demonstrate that selective photoinhibition of these neurons affects mystacial pad activity, with minimal effects on breathing. These effects are altered by the type of anesthetic employed and also between anesthetized and conscious states. The population of 7n-projecting preBötC neurons we transduced consisted of both excitatory and inhibitory neurons that also send collaterals to multiple brainstem nuclei involved with the regulation of autonomic activity. We show that modulation of subgroups of preBötC neurons, based on their axonal projections, is a useful strategy to improve our understanding of the mechanisms that coordinate and integrate breathing with different motor and physiological behaviors. This is of fundamental importance, given that abnormal respiratory modulation of autonomic activity and orofacial behaviors have been associated with the development and progression of diseases.


While breathing seems to come easy, it is a complex process in which many muscles coordinate to allow air to flow into the lungs. These muscles also control the flow of air we breathe out to allow us to talk, sing, eat, or drink. The brain circuits that control these muscles, can also influence other parts of the brain. The preBötzinger Complex, which is a key region of brainstem circuits that generate and control breathing, contains neurons that also project widely, connecting to other regions of the brain. This helps to modulate the sense of smell, emotional state, heart rate, and even blood pressure. Understanding how the preBötzinger Complex is organized can untangle how breathing can influence these other processes. Melo et al. wanted to learn whether they could manipulate the activity of a subgroup of preBötzinger Complex neurons that project into the facial nucleus ­ a region of the brain that controls the muscles of the face when we breathe ­ without affecting breathing. If this can be done, it might also be possible to affect blood pressure by manipulating selective preBötzinger neurons, and thus the development of hypertension, without having any impact on breathing. To test this hypothesis, Melo et al. used rats in which the activation of preBötzinger Complex neurons that project into the facial nucleus was blocked. This decreased the activity of the muscles around the nose with hardly any effect on breathing. Melo et al. also found that the state of consciousness of the rat (anesthetized or conscious) could affect how preBötzinger Complex neurons control these muscles. Melo et al. also observed that preBötzinger Complex neurons projecting into the facial nucleus had projections into many other regions in the brainstem. This might help to the coordinate respiratory, cardiovascular, orofacial, and potentially other physiological functions. The findings of Melo et al. set a technical foundation for exploring the influence of specific subgroups of preBötzinger Complex neurons on respiratory modulation of other physiological activities, including blood pressure and heart rate and in conditions, such as hypertension and heart failure. More broadly, most brain regions contain complex and heterogeneous groups of neurons and the strategy validated by Melo et. al. could be applied to unravel other brain-function relationships.


Asunto(s)
Núcleo Motor del Nervio Facial , Ratas , Animales , Centro Respiratorio , Respiración , Neuronas Motoras , Tronco Encefálico
6.
Neurosci Lett ; 787: 136817, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35905886

RESUMEN

Interruption of the activity of neurons in the commissural portion of the nucleus of the solitary tract (cNTS) decreases blood pressure (BP) in experimental models of hypertension, such as the spontaneously hypertensive (SH) rat. To examine whether PHOX2B expressing cNTS neurons are involved in maintaining the elevated BP, we used replication-deficient viruses with a modified Phox2 binding site promoter to express the inhibitory chemogenetic allatostatin receptor or green fluorescent protein in the cNTS. Following administration of allatostatin, we observed a depressor and bradycardic response in anesthetized SH rats that expressed the allatostatin receptor. Injection of allatostatin did not affect BP or heart rate (HR) in control SH rats expressing green fluorescent protein in the cNTS. Immunohistochemistry showed that the majority of transduced cNTS neurons were PHOX2B-immunoreactive and some also expressed tyrosine hydroxylase. We conclude that in anesthetized SH rat, the Phox2B expressing cNTS neurons maintain elevated BP.


Asunto(s)
Neuronas , Núcleo Solitario , Animales , Presión Sanguínea , Proteínas Fluorescentes Verdes/metabolismo , Frecuencia Cardíaca , Neuronas/metabolismo , Ratas , Ratas Endogámicas SHR , Núcleo Solitario/metabolismo , Factores de Transcripción/metabolismo
7.
J Comp Neurol ; 530(17): 3072-3103, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35988033

RESUMEN

Anatomical tracing studies examining the vagal system can conflate details of sensory afferent and motor efferent neurons. Here, we used a serotype of adeno-associated virus that transports retrogradely and exhibits selective tropism for vagal afferents, to map their soma location and central termination sites within the nucleus of the solitary tract (NTS). We examined the vagal sensory afferents innervating the trachea, duodenum, stomach, or heart, and in some animals, from two organs concurrently. We observed no obvious somatotopy in the somata distribution within the nodose ganglion. The central termination patterns of afferents from different organs within the NTS overlap substantially. Convergence of vagal afferent inputs from different organs onto single NTS neurons is observed. Abdominal and thoracic afferents terminate throughout the NTS, including in the rostral NTS, where the 7th cranial nerve inputs are known to synapse. To address whether the axonal labeling produced by viral transduction is so widespread because it fills axons traveling to their targets, and not just terminal fields, we labeled pre and postsynaptic elements of vagal afferents in the NTS . Vagal afferents form multiple putative synapses as they course through the NTS, with each vagal afferent neuron distributing sensory signals to multiple second-order NTS neurons. We observe little selectivity between vagal afferents from different visceral targets and NTS neurons with common neurochemical phenotypes, with afferents from different organs making close appositions with the same NTS neuron. We conclude that specific viscerosensory information is distributed widely within the NTS and that the coding of this input is probably determined by the intrinsic properties and projections of the second-order neuron.


Asunto(s)
Núcleo Solitario , Nervio Vago , Animales , Neuronas Motoras , Neuronas Aferentes/fisiología , Ganglio Nudoso , Ratas , Núcleo Solitario/fisiología , Nervio Vago/fisiología
8.
Cell Rep ; 32(11): 108139, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937120

RESUMEN

Chemogenetics enables manipulation of neuronal activity in experimental animals. While providing information about the transduced neuron expressing a ligand-activated molecule, chemogenetics does not provide understanding about the antecedent circuit that drives that neuron's activity. For current approaches, this is not feasible, because the activating molecules are not genetically encoded. The insect allatostatin/allatostatin receptor system, a highly specific, powerful inhibitory chemogenetic approach, has this advantage, because the ligand, being a peptide, is genetically encoded. We developed viral vector-based systems to express biologically active allatostatin in neurons in vivo and allatostatin receptors in subpopulations of postsynaptic neurons. We demonstrate that activity-dependent release of allatostatin induces inhibition of allatostatin receptor-expressing neurons. We validate the approach in the vagal viscerosensory system where inhibitory, rather than the usual excitatory, viscerosensory input leads to sustained decreases in baroreceptor reflex sensitivity and bodyweight.


Asunto(s)
Red Nerviosa/fisiología , Neuronas/fisiología , Secuencia de Aminoácidos , Animales , Presión Sanguínea , Peso Corporal , Células CHO , Cricetulus , Fenómenos Electrofisiológicos , Células HEK293 , Proteínas de Homeodominio , Homeostasis , Humanos , Neuronas Aferentes/fisiología , Neuropéptidos/química , Neuropéptidos/metabolismo , Ratas Endogámicas SHR , Ratas Sprague-Dawley , Ratas Transgénicas , Receptores de Superficie Celular/metabolismo , Núcleo Solitario/fisiología , Sinapsis/metabolismo , Transgenes , Nervio Vago/fisiología
9.
Elife ; 92020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32538785

RESUMEN

Heart rate and blood pressure oscillate in phase with respiratory activity. A component of these oscillations is generated centrally, with respiratory neurons entraining the activity of pre-sympathetic and parasympathetic cardiovascular neurons. Using a combination of optogenetic inhibition and excitation in vivo and in situ in rats, as well as neuronal tracing, we demonstrate that preBötzinger Complex (preBötC) neurons, which form the kernel for inspiratory rhythm generation, directly modulate cardiovascular activity. Specifically, inhibitory preBötC neurons modulate cardiac parasympathetic neuron activity whilst excitatory preBötC neurons modulate sympathetic vasomotor neuron activity, generating heart rate and blood pressure oscillations in phase with respiration. Our data reveal yet more functions entrained to the activity of the preBötC, with a role in generating cardiorespiratory oscillations. The findings have implications for cardiovascular pathologies, such as hypertension and heart failure, where respiratory entrainment of heart rate is diminished and respiratory entrainment of blood pressure exaggerated.


Asunto(s)
Presión Sanguínea , Frecuencia Cardíaca , Neuronas/fisiología , Centro Respiratorio/fisiología , Potenciales de Acción , Animales , Canales de Cloruro/fisiología , Potenciales Postsinápticos Excitadores , Masculino , Bulbo Raquídeo/fisiología , Optogenética , Ratas , Ratas Sprague-Dawley , Respiración
10.
Biol Sex Differ ; 9(1): 14, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29625592

RESUMEN

BACKGROUND: Over 100 mammalian G protein-coupled receptors are yet to be matched with endogenous ligands; these so-called orphans are prospective drug targets for the treatment of disease. GPR37L1 is one such orphan, abundant in the brain and detectable as mRNA in the heart and kidney. GPR37L1 ablation was reported to cause hypertension and left ventricular hypertrophy, and thus, we sought to further define the role of GPR37L1 in blood pressure homeostasis. METHODS: We investigated the cardiovascular effects of GPR37L1 using wild-type (GPR37L1wt/wt) and null (GPR37L1KO/KO) mice established on a C57BL/6J background, both under baseline conditions and during AngII infusion. We profiled GPR37L1 tissue expression, examining the endogenous receptor by immunoblotting and a ß-galactosidase reporter mouse by immunohistochemistry. RESULTS: GPR37L1 protein was abundant in the brain but not detectable in the heart and kidney. We measured blood pressure in GPR37L1wt/wt and GPR37L1KO/KO mice and found that deletion of GPR37L1 causes a female-specific increase in systolic, diastolic, and mean arterial pressures. When challenged with short-term AngII infusion, only male GPR37L1KO/KO mice developed exacerbated left ventricular hypertrophy and evidence of heart failure, while the female GPR37L1KO/KO mice were protected from cardiac fibrosis. CONCLUSIONS: Despite its absence in the heart and kidney, GPR37L1 regulates baseline blood pressure in female mice and is crucial for cardiovascular compensatory responses in males. The expression of GPR37L1 in the brain, yet absence from peripheral cardiovascular tissues, suggests this orphan receptor is a hitherto unknown contributor to central cardiovascular control.


Asunto(s)
Presión Sanguínea , Receptores Acoplados a Proteínas G/fisiología , Animales , Encéfalo/metabolismo , Femenino , Fibrosis , Riñón/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Caracteres Sexuales
11.
Cell Metab ; 25(3): 739-748, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28215844

RESUMEN

The etiology of hypertension, the world's biggest killer, remains poorly understood, with treatments targeting the established symptom, not the cause. The development of hypertension involves increased sympathetic nerve activity that, in experimental hypertension, may be driven by excessive respiratory modulation. Using selective viral and cell lesion techniques, we identify adrenergic C1 neurons in the medulla oblongata as critical for respiratory-sympathetic entrainment and the development of experimental hypertension. We also show that a cohort of young, normotensive humans, selected for an exaggerated blood pressure response to exercise and thus increased hypertension risk, has enhanced respiratory-related blood pressure fluctuations. These studies pinpoint a specific neuronal target for ameliorating excessive sympathetic activity during the developmental phase of hypertension and identify a group of pre-hypertensive subjects that would benefit from targeting these cells.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Respiración , Envejecimiento/fisiología , Animales , Neuronas/fisiología , Ratas Endogámicas SHR , Sistema Nervioso Simpático/fisiopatología , Sinapsis/fisiología
12.
Hypertension ; 62(5): 866-71, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24001896

RESUMEN

Hypertension contributes to multiple forms of cardiovascular disease and thus morbidity and mortality. The mechanisms inducing hypertension remain unclear although the involvement of homeostatic systems, such as the renin-angiotensin and sympathetic nervous systems, is established. A pivotal role of the angiotensin type 1 receptor in the proximal tubule of the kidney for the development of experimental hypertension is established. Yet, other systems are involved. This study tests whether the expression of angiotensin type 1A receptors in catecholaminergic cells contributes to hypertension development. Using a Cre-lox approach, we deleted the angiotensin type 1A receptor from all catecholaminergic cells. This deletion did not alter basal metabolism or blood pressure but delayed the onset of angiotensin-dependent hypertension and reduced the maximal response. Cardiac hypertrophy was also reduced. The knockout mice showed attenuated activation of the sympathetic nervous system during angiotensin II infusion as measured by spectral analysis of the blood pressure. Increased reactive oxygen species production was observed in forebrain regions, including the subfornical organ, of the knockout mouse but was markedly reduced in the rostral ventrolateral medulla. These studies demonstrate that stimulation of the angiotensin type 1A receptor on catecholaminergic cells is required for the full development of angiotensin-dependent hypertension and support an important role for the sympathetic nervous system in this model.


Asunto(s)
Presión Sanguínea/fisiología , Cardiomegalia/metabolismo , Catecolaminas/metabolismo , Hipertensión/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina II , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/fisiopatología , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/genética , Órgano Subfornical/efectos de los fármacos , Órgano Subfornical/metabolismo , Órgano Subfornical/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología
13.
Cardiovasc Res ; 100(2): 181-91, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23887975

RESUMEN

AIMS: The nucleus of the solitary tract (NTS) is important for cardiovascular regulation and contains angiotensin type 1A (AT1A) receptors. To assess its function, we examined the effect of expressing in AT1A receptors in the NTS of mice lacking these receptors. METHODS AND RESULTS: Bilateral microinjections of lentivirus expressing AT1A receptors (AT1Av mice, n = 6) or green fluorescent protein (GFPv, n = 8, control) under the control of the PRSx8 promotor were made into the NTS of AT1A receptors null mice (AT1A(-/-)). Telemetry devices recorded blood pressure (BP), heart rate (HR), and locomotor activity. Expression of AT1A receptors in the NTS increased BP by 11.2 ± 4 mmHg (P < 0.05) at 2 and 3 weeks, whereas GFPv mice remained at pre-injection BP. Ganglion blockade reduced BP to similar levels pre- and post-transfection in GFPv and AT1Av mice. Greater pressor responses to cage-switch stress were observed following AT1A receptors expression (+18 ± 2 mmHg pre- to +24 ± 2 mmHg post-virus, P < 0.05) with similar stress-induced pressor responses pre- and post-virus in GFPv mice. Pressor responses to restraint stress pre- and post-virus were similar in AT1Av but were 20% less post-GFPv (P < 0.001). The lack of attenuation in BP to restraint was associated with four-fold greater Fos-expression in AT1A receptors mice. AT1A receptors expression in the NTS did not alter baroreflex gain differently between groups. CONCLUSION: The results suggest that transfection of AT1A receptors on neurons in the NTS elevates BP independent of the SNS and pressor responses to aversive stimuli are associated with greater Fos-expression in forebrain regions. This study suggests a novel mechanism by which the NTS may modulate MAP in the long-term via AT1A receptors.


Asunto(s)
Presión Arterial/fisiología , Frecuencia Cardíaca/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Núcleo Solitario/fisiología , Animales , Autorradiografía , Barorreflejo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Tartrato de Pentolinio/farmacología , Proteínas Proto-Oncogénicas c-fos/análisis , Estrés Psicológico/fisiopatología
14.
Cardiovasc Res ; 96(2): 330-9, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22869618

RESUMEN

AIMS: The caudal ventrolateral medulla (CVLM) is important for autonomic regulation and is rich in angiotensin II type 1A receptors (AT(1A)R). To determine their function, we examined whether the expression of AT(1A)R in the CVLM of mice lacking AT(1A)R (AT(1A)(-/-)) alters baroreflex sensitivity and cardiovascular responses to stress. METHODS AND RESULTS: Bilateral microinjections into the CVLM of AT(1A)(-/-) mice of lentivirus with the phox-2 selective promoter (PRSx8) were made to express either AT(1A)R (Lv-PRSx8-AT(1A)) or green fluorescent protein (Lv-PRSx8-GFP) as a control. Radiotelemetry was used to record mean arterial pressure (MAP), heart rate (HR), and locomotor activity. Following injection of Lv-PRSx8-GFP, robust neuronal expression of GFP was observed with ∼60% of the GFP-positive cells also expressing the catecholamine-synthetic enzyme tyrosine hydroxylase. After 5 weeks, there were no differences in MAP or HR between groups, but the Lv-PRSx8-AT(1A)- injected mice showed reduced baroreflex sensitivity (-25%, P = 0.003) and attenuated pressor responses to cage-switch and restraint stress compared with the Lv-PRSx8-GFP-injected mice. Reduced MAP mid-frequency power during cage-switch stress reflected attenuated sympathetic activation (Pgroup × stress = 0.04). Fos-immunohistochemistry indicated greater activation of forebrain and hypothalamic neurons in the Lv-PRSx8-AT(1A) mice compared with the control. CONCLUSION: The expression of AT(1A)R in CVLM neurons, including A1 neurons, while having little influence on the basal blood pressure or HR, may play a tonic role in inhibiting cardiac vagal baroreflex sensitivity. However, they strongly facilitate the forebrain response to aversive stress, yet reduce the pressor response presumably through greater sympatho-inhibition. These findings outline novel and specific roles for angiotensin II in the CVLM in autonomic regulation.


Asunto(s)
Barorreflejo , Bulbo Raquídeo/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Estrés Psicológico , Animales , Autorradiografía , Presión Sanguínea , Técnicas de Transferencia de Gen , Frecuencia Cardíaca , Inmunohistoquímica , Lentivirus , Masculino , Ratones , Actividad Motora , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Restricción Física , Transgenes
15.
Hypertension ; 56(1): 143-50, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20458002

RESUMEN

In adult mice we determined whether expression of angiotensin II (Ang II) type 1A receptors (AT(1A)Rs) in C1 neurons mediates the excitation of the rostral ventrolateral medulla (RVLM) by Ang II. Blood pressure, heart rate, and sympathetic nerve activity were measured in anesthetized, artificially ventilated wild-type (n=15) and AT(1A)R knockout (AT(1A)(-/-); n=9) mice. Microinjection of Ang II (50 nL of 0.1 to 1.0 mmol/L) into the RVLM induced a dose-related, sympathetically mediated pressor response (maximum of 17+/-2 mm Hg) in wild-type mice. These microinjections had no effect in AT(1A)(-/-) mice. Endogenous AT(1)Rs occur on catecholaminergic C1 neurons in the RVLM. We induced AT(1A)R or green fluorescent protein expression in C1 neurons of AT(1A)(-/-) mice through bilateral microinjection of replication-deficient lentiviruses, with transgene expression under the control of a phox2 transcription factor binding promoter (PRSx8) (Lv-PRSx8-AT(1A), n=10, and Lv-PRSx8-GFP, n=5). Transgene expression was observed in a significant proportion of RVLM C1 neurons. In anesthetized Lv-PRSx8-AT(1A) injected mice, unilateral RVLM microinjection of Ang II (50 nL of 1 mmol/L) increased blood pressure (17+/-4 mm Hg) and sympathetic nerve activity (155+/-32%). No response to Ang II occurred in Lv-PRSx8-GFP microinjected mice. These results show that Ang II-mediated excitation of RVLM neurons in adult mice depends on the AT(1A)R with little or no effect of type 1B or 2 receptors. Expression of the AT(1A)R predominantly in C1 catecholamine neurons restores the response to Ang II in the AT(1A)(-/-) mouse and demonstrates that these neurons are sympathoexcitatory in the mouse.


Asunto(s)
Angiotensina II/farmacología , Presión Sanguínea/fisiología , Bulbo Raquídeo/efectos de los fármacos , Neuronas/metabolismo , Receptor de Angiotensina Tipo 1/biosíntesis , Sistema Nervioso Simpático/efectos de los fármacos , Animales , Autorradiografía , Inmunohistoquímica , Bulbo Raquídeo/fisiología , Ratones , Ratones Noqueados , Sistema Nervioso Simpático/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA