Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 9(6)2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38349753

RESUMO

Glucose homeostasis is achieved via complex interactions between the endocrine pancreas and other peripheral tissues and glucoregulatory neurocircuits in the brain that remain incompletely defined. Within the brain, neurons in the hypothalamus appear to play a particularly important role. Consistent with this notion, we report evidence that (pro)renin receptor (PRR) signaling within a subset of tyrosine hydroxylase (TH) neurons located in the hypothalamic paraventricular nucleus (PVNTH neurons) is a physiological determinant of the defended blood glucose level. Specifically, we demonstrate that PRR deletion from PVNTH neurons restores normal glucose homeostasis in mice with diet-induced obesity (DIO). Conversely, chemogenetic inhibition of PVNTH neurons mimics the deleterious effect of DIO on glucose. Combined with our finding that PRR activation inhibits PVNTH neurons, these findings suggest that, in mice, (a) PVNTH neurons play a physiological role in glucose homeostasis, (b) PRR activation impairs glucose homeostasis by inhibiting these neurons, and (c) this mechanism plays a causal role in obesity-associated metabolic impairment.


Assuntos
Glucose , Receptor de Pró-Renina , Animais , Camundongos , Glucose/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Obesidade/complicações , Obesidade/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
2.
Am J Physiol Cell Physiol ; 325(1): C141-C154, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37273237

RESUMO

The regulation of plasma glucose levels is a complex and multifactorial process involving a network of receptors and signaling pathways across numerous organs that act in concert to ensure homeostasis. However, much about the mechanisms and pathways by which the brain regulates glycemic homeostasis remains poorly understood. Understanding the precise mechanisms and circuits employed by the central nervous system to control glucose is critical to resolving the diabetes epidemic. The hypothalamus, a key integrative center within the central nervous system, has recently emerged as a critical site in the regulation of glucose homeostasis. Here, we review the current understanding of the role of the hypothalamus in regulating glucose homeostasis, with an emphasis on the paraventricular nucleus, the arcuate nucleus, the ventromedial hypothalamus, and lateral hypothalamus. In particular, we highlight the emerging role of the brain renin-angiotensin system in the hypothalamus in regulating energy expenditure and metabolic rate, as well as its potential importance in the regulation of glucose homeostasis.


Assuntos
Hipotálamo , Sistema Renina-Angiotensina , Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Hipotálamo/metabolismo , Humanos , Animais
3.
Biomolecules ; 12(9)2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-36139008

RESUMO

The brain renin-angiotensin system plays important roles in blood pressure and cardiovascular regulation. There are two isoforms of prorenin in the brain: the classic secreted form (prorenin/sREN) encoded by renin-a, and an intracellular form (icREN) encoded by renin-b. Emerging evidence indicates the importance of renin-b in cardiovascular and metabolic regulation. However, the role of endogenous brain prorenin in the development of salt-sensitive hypertension remains undefined. In this study, we test the hypothesis that renin-a produced locally in the brain contributes to the pathogenesis of hypertension. Using RNAscope, we report for the first time that renin mRNA is expressed in several regions of the brain, including the subfornical organ (SFO), the paraventricular nucleus of the hypothalamus (PVN), and the brainstem, where it is found in glutamatergic, GABAergic, cholinergic, and tyrosine hydroxylase-positive neurons. Notably, we found that renin mRNA was significantly elevated in the SFO and PVN in a mouse model of DOCA-salt-induced hypertension. To examine the functional importance of renin-a in the SFO, we selectively ablated renin-a in the SFO in renin-a-floxed mice using a Cre-lox strategy. Importantly, renin-a ablation in the SFO attenuated the maintenance of DOCA-salt-induced hypertension and improved autonomic function without affecting fluid or sodium intake. Molecularly, ablation of renin-a prevented the DOCA-salt-induced elevation in NADPH oxidase 2 (NOX2) in the SFO without affecting NOX4 or angiotensin II type 1 and 2 receptors. Collectively, our findings demonstrate that endogenous renin-a within the SFO is important for the pathogenesis of salt-sensitive hypertension.


Assuntos
Acetato de Desoxicorticosterona , Hipertensão , Sódio na Dieta , Órgão Subfornical , Angiotensina II , Animais , Colinérgicos , Hipertensão/genética , Hipertensão/metabolismo , Camundongos , NADPH Oxidase 2 , RNA Mensageiro/metabolismo , Renina/genética , Cloreto de Sódio , Sódio na Dieta/efeitos adversos , Órgão Subfornical/metabolismo , Tirosina 3-Mono-Oxigenase
4.
Physiol Rep ; 9(3): e14753, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33587331

RESUMO

Chlorisondamine (CSD) has been used to assess the neurogenic contribution to blood pressure (BP) and vasomotor sympathetic tone in animal models. It is assumed that the reduction in BP following CSD administration is associated to decreases in cardiac output (CO) and peripheral resistance, reflecting cardiac and vasomotor sympathetic tone, respectively. Surprisingly, this has not been characterized experimentally in mice, despite the extensive use of this animal model in cardiovascular research. We hypothesize that a specific dose of CSD can selectively block the sympathetic vasomotor tone. To test this hypothesis, we evaluated the effects of different doses of CSD (intraperitoneal) on BP and heart rate (HR) using telemetry, and on CO using echocardiography. BP and HR in normotensive C57Bl/6J mice reduced to a similar extent by all CSD doses tested (1-6 mg/kg). CSD at 6 mg/kg also reduced CO without affecting left ventricular stroke volume or fractional shortening. On the other hand, lower doses of CSD (1 and 2 mg/kg) produced significantly larger BP and HR reductions in DOCA-salt-induced hypertensive mice, indicating a greater neurogenic BP response. In addition, all doses of CSD reduced CO in hypertensive mice. Our data suggest that the BP response to CSD in mice likely reflects reduced CO and vasomotor sympathetic tone. We conclude that CSD can be used to assess the neurogenic contribution to BP in mice but may not be appropriate for specifically estimating vasomotor sympathetic tone.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Sistema Cardiovascular/inervação , Clorisondamina/farmacologia , Hipertensão/fisiopatologia , Sistema Nervoso Simpático/efeitos dos fármacos , Simpatolíticos/farmacologia , Animais , Débito Cardíaco/efeitos dos fármacos , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/etiologia , Masculino , Camundongos Endogâmicos C57BL , Cloreto de Sódio na Dieta , Sistema Nervoso Simpático/fisiopatologia , Sistema Vasomotor/efeitos dos fármacos , Sistema Vasomotor/fisiopatologia
5.
Front Physiol ; 11: 606811, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329061

RESUMO

Overactivation of the renin-angiotensin system (RAS) - a central physiological pathway involved in controlling blood pressure (BP) - leads to hypertension. It is now well-recognized that the central nervous system (CNS) has its own local RAS, and the majority of its components are known to be expressed in the brain. In physiological and pathological states, the (pro)renin receptor (PRR), a novel component of the brain RAS, plays a key role in the formation of angiotensin II (Ang II) and also mediates Ang II-independent PRR signaling. A recent study reported that neuronal PRR activation is a novel mechanism for cardiovascular and metabolic regulation in obesity and diabetes. Expression of the PRR is increased in cardiovascular regulatory nuclei in hypertensive (HTN) animal models and plays an important role in BP regulation in the CNS. To determine the clinical significance of the brain PRR in human hypertension, we investigated whether the PRR is expressed and regulated in the paraventricular nucleus of the hypothalamus (PVN) and rostral ventrolateral medulla (RVLM) - two key cardiovascular regulatory nuclei - in postmortem brain samples of normotensive (NTN) and HTN humans. Here, we report that the PRR is expressed in neurons, but not astrocytes, of the human PVN and RVLM. Notably, PRR immunoreactivity was significantly increased in both the PVN and RVLM of HTN subjects. In addition, PVN-PRR immunoreactivity was positively correlated with systolic BP (sBP) and showed a tendency toward correlation with age but not body mass index (BMI). Collectively, our data provide clinical evidence that the PRR in the PVN and RVLM may be a key molecular player in the neural regulation of BP and cardiovascular and metabolic function in humans.

6.
Am J Physiol Endocrinol Metab ; 318(5): E765-E778, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32228320

RESUMO

We report here that the neuronal (pro)renin receptor (PRR), a key component of the brain renin-angiotensin system (RAS), plays a critical role in the central regulation of high-fat-diet (HFD)-induced metabolic pathophysiology. The neuronal PRR is known to mediate formation of the majority of angiotensin (ANG) II, a key bioactive peptide of the RAS, in the central nervous system and to regulate blood pressure and cardiovascular function. However, little is known about neuronal PRR function in overnutrition-related metabolic physiology. Here, we show that PRR deletion in neurons reduces blood pressure, neurogenic pressor activity, and fasting blood glucose and improves glucose tolerance without affecting food intake or body weight following a 16-wk HFD. Mechanistically, we found that a HFD increases levels of the PRR ligand (pro)renin in the circulation and hypothalamus and of ANG II in the hypothalamus, indicating activation of the brain RAS. Importantly, PRR deletion in neurons reduced astrogliosis and activation of the astrocytic NF-κB p65 (RelA) in the arcuate nucleus and the ventromedial nucleus of the hypothalamus. Collectively, our findings indicate that the neuronal PRR plays essential roles in overnutrition-related metabolic pathophysiology.


Assuntos
Astrócitos/metabolismo , Glicemia/metabolismo , Pressão Sanguínea/fisiologia , Hipotálamo/metabolismo , Inflamação/metabolismo , Neurônios/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Peso Corporal/fisiologia , Dieta Hiperlipídica , Ingestão de Alimentos/fisiologia , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/genética , Renina/metabolismo , Receptor de Pró-Renina
7.
Am J Physiol Heart Circ Physiol ; 317(3): H496-H504, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31274353

RESUMO

Accumulating evidence supports that the brain renin-angiotensin system (RAS), including prorenin (PR) and its receptor (PRR), two newly discovered RAS players, contribute to sympathoexcitation in salt-sensitive hypertension. Still, whether PR also contributed to elevated circulating levels of neurohormones such as vasopressin (VP) during salt-sensitive hypertension, and if so, what are the precise underlying mechanisms, remains to be determined. To address these questions, we obtained patch-clamp recordings from hypothalamic magnocellular neurosecretory neurons (MNNs) that synthesize the neurohormones oxytocin and VP in acute hypothalamic slices obtained from sham and deoxycorticosterone acetate (DOCA)-salt-treated hypertensive rats. We found that focal application of PR markedly increased membrane excitability and firing responses in MNNs of DOCA-salt, compared with sham rats. This effect included a shorter latency to spike initiation and increased numbers of spikes in response to depolarizing stimuli and was mediated by a more robust inhibition of A-type K+ channels in DOCA-salt compared with sham rats. On the other hand, the afterhyperpolarizing potential mediated by the activation of Ca2+-dependent K+ channel was not affected by PR. mRNA expression of PRR, VP, and the Kv4.3 K+ channel subunit in the supraoptic nucleus of DOCA-salt hypertensive rats was increased compared with sham rats. Finally, we report a significant decrease of plasma VP levels in neuron-selective PRR knockdown mice treated with DOCA-salt, compared with wild-type DOCA-salt-treated mice. Together, these results support that activation of PRR contributes to increased excitability and firing discharge of MNNs and increased plasma levels of VP in DOCA-salt hypertension.NEW & NOTEWORTHY Our studies support that prorenin (PR) and its receptor (PRR) within the hypothalamus contribute to elevated plasma vasopressin levels in deoxycorticosterone acetate-salt hypertension, in part because of an exacerbated effect of PR on magnocellular neurosecretory neuron excitability; Moreover, our study implicates A-type K+ channels as key underlying molecular targets mediating these effects. Thus, PR/PRR stands as a novel therapeutic target for the treatment of neurohumoral activation in salt-sensitive hypertension.


Assuntos
Pressão Sanguínea , Hipertensão/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Sistema Renina-Angiotensina , Renina/metabolismo , Vasopressinas/sangue , Animais , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , Hipertensão/sangue , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Hipotálamo/fisiopatologia , Masculino , Potenciais da Membrana , Camundongos Knockout , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/metabolismo , Ratos Wistar , Tempo de Reação , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Canais de Potássio Shaw/genética , Canais de Potássio Shaw/metabolismo , Cloreto de Sódio na Dieta , Fatores de Tempo , Regulação para Cima
8.
Am J Physiol Heart Circ Physiol ; 316(6): H1389-H1405, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30925093

RESUMO

Activation of the brain renin-angiotensin system (RAS) is a pivotal step in the pathogenesis of hypertension. The paraventricular nucleus (PVN) of the hypothalamus is a critical part of the angiotensinergic sympatho-excitatory neuronal network involved in neural control of blood pressure and hypertension. However, the importance of the PVN (pro)renin receptor (PVN-PRR)-a key component of the brain RAS-in hypertension development has not been examined. In this study, we investigated the involvement and mechanisms of the PVN-PRR in DOCA-salt-induced hypertension, a mouse model of hypertension. Using nanoinjection of adeno-associated virus-mediated Cre recombinase expression to knock down the PRR specifically in the PVN, we report here that PVN-PRR knockdown attenuated the enhanced blood pressure and sympathetic tone associated with hypertension. Mechanistically, we found that PVN-PRR knockdown was associated with reduced activation of ERK (extracellular signal-regulated kinase)-1/2 in the PVN and rostral ventrolateral medulla during hypertension. In addition, using the genetically encoded Ca2+ biosensor GCaMP6 to monitor Ca2+-signaling events in the neurons of PVN brain slices, we identified a reduction in angiotensin II type 1 receptor-mediated Ca2+ activity as part of the mechanism by which PVN-PRR knockdown attenuates hypertension. Our study demonstrates an essential role of the PRR in PVN neurons in hypertension through regulation of ERK1/2 activation and angiotensin II type 1 receptor-mediated Ca2+ activity. NEW & NOTEWORTHY PRR knockdown in PVN neurons attenuates the development of DOCA-salt hypertension and autonomic dysfunction through a decrease in ERK1/2 activation in the PVN and RVLM during hypertension. In addition, PRR knockdown reduced AT1aR expression and AT1R-mediated calcium activity during hypertension. Furthermore, we characterized the neuronal targeting specificity of AAV serotype 2 in the mouse PVN and validated the advantages of the genetically encoded calcium biosensor GCaMP6 in visualizing neuronal calcium activity in the PVN.


Assuntos
Pressão Sanguínea , Sinalização do Cálcio , Hipertensão/prevenção & controle , Neurônios/enzimologia , Núcleo Hipotalâmico Paraventricular/enzimologia , ATPases Translocadoras de Prótons/deficiência , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Superfície Celular/deficiência , Animais , Sistema Nervoso Autônomo/metabolismo , Sistema Nervoso Autônomo/fisiopatologia , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Hipertensão/enzimologia , Hipertensão/genética , Hipertensão/fisiopatologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Fosforilação , ATPases Translocadoras de Prótons/genética , Receptor Tipo 1 de Angiotensina/genética , Receptores de Superfície Celular/genética , Receptor de Pró-Renina
9.
Am J Physiol Heart Circ Physiol ; 314(3): H580-H592, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29350998

RESUMO

Despite advances in antihypertensive therapeutics, at least 15-20% of hypertensive patients have resistant hypertension through mechanisms that remain poorly understood. In this study, we provide a new mechanism for the regulation of blood pressure (BP) in the central nervous system (CNS) by the (pro)renin receptor (PRR), a recently identified component of the renin-angiotensin system that mediates ANG II formation in the CNS. Although PRR also mediates ANG II-independent signaling, the importance of these pathways in BP regulation is unknown. Here, we developed a unique transgenic mouse model overexpressing human PRR (hPRR) specifically in neurons (Syn-hPRR). Intracerebroventricular infusion of human prorenin caused increased BP in Syn-hPRR mice. This BP response was attenuated by a NADPH oxidase (NOX) inhibitor but not by antihypertensive agents that target the renin-angiotensin system. Using a brain-targeted genetic knockdown approach, we found that NOX4 was the key isoform responsible for the prorenin-induced elevation of BP in Syn-hPRR mice. Moreover, inhibition of ERK significantly attenuated the increase in NOX activity and BP induced by human prorenin. Collectively, our findings indicate that an ANG II-independent, PRR-mediated signaling pathway regulates BP in the CNS by a PRR-ERK-NOX4 mechanism. NEW & NOTEWORTHY This study characterizes a new transgenic mouse model with overexpression of the human (pro)renin receptor in neurons and demonstrated a novel angiotensin II-independent mechanism mediated by human prorenin and the (pro)renin receptor in the central regulation of blood pressure.


Assuntos
Angiotensina II , Pressão Sanguínea , Sistema Nervoso Central/enzimologia , Hipertensão/induzido quimicamente , Hipertensão/enzimologia , Neurônios/enzimologia , Receptores de Superfície Celular/metabolismo , Sistema Renina-Angiotensina , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Hipertensão/genética , Hipertensão/fisiopatologia , Infusões Intraventriculares , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo , Neurônios/efeitos dos fármacos , Regiões Promotoras Genéticas , Receptores de Superfície Celular/genética , Renina/administração & dosagem , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/genética , Transdução de Sinais , Sinapsinas/genética , Regulação para Cima , ATPases Vacuolares Próton-Translocadoras/genética
10.
Am J Physiol Heart Circ Physiol ; 314(4): H796-H804, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351470

RESUMO

The central nervous system plays an important role in essential hypertension in humans and in animal models of hypertension through modulation of sympathetic activity and Na+ and body fluid homeostasis. Data from animal models of hypertension suggest that the renin-angiotensin system in the subfornical organ (SFO) of the brain is critical for hypertension development. We recently reported that the brain (pro)renin receptor (PRR) is a novel component of the brain renin-angiotensin system and could be a key initiator of the pathogenesis of hypertension. Here, we examined the expression level and cellular distribution of PRR in the SFO of postmortem human brains to assess its association with the pathogenesis of human hypertension. Postmortem SFO tissues were collected from hypertensive and normotensive human subjects. Immunolabeling for the PRR and a retrospective analysis of clinical data were performed. We found that human PRR was prominently expressed in most neurons and microglia, but not in astrocytes, in the SFO. Importantly, PRR levels in the SFO were elevated in hypertensive subjects. Moreover, PRR immunoreactivity was significantly correlated with systolic blood pressure but not body weight, age, or diastolic blood pressure. Interestingly, this correlation was independent of antihypertensive drug therapy. Our data indicate that PRR in the SFO may be a key molecular player in the pathogenesis of human hypertension and, as such, could be an important focus of efforts to understand the neurogenic origin of hypertension. NEW & NOTEWORTHY This study provides evidence that, in the subfornical organ of the human brain, the (pro)renin receptor is expressed in neurons and microglia cells but not in astrocytes. More importantly, (pro)renin receptor immunoreactivity in the subfornical organ is increased in hypertensive humans and is significantly correlated with systolic blood pressure.


Assuntos
Hipertensão/enzimologia , Receptores de Superfície Celular/análise , Órgão Subfornical/enzimologia , ATPases Vacuolares Próton-Translocadoras/análise , Idoso , Autopsia , Pressão Sanguínea , Feminino , Humanos , Hipertensão/diagnóstico , Hipertensão/fisiopatologia , Imuno-Histoquímica , Masculino , Microglia/enzimologia , Pessoa de Meia-Idade , Neurônios/enzimologia , Estudos Retrospectivos , Órgão Subfornical/fisiopatologia , Regulação para Cima
11.
Hypertension ; 65(2): 352-61, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25421983

RESUMO

We previously reported that binding of prorenin to the (pro)renin receptor (PRR) plays a major role in brain angiotensin II formation and the development of deoxycorticosterone acetate (DOCA)-salt hypertension. Here, we designed and developed an antagonistic peptide, PRO20, to block prorenin binding to the PRR. Fluorescently labeled PRO20 bound to both mouse and human brain tissues with dissociation constants of 4.4 and 1.8 nmol/L, respectively. This binding was blocked by coincubation with prorenin and was diminished in brains of neuron-specific PRR-knockout mice, indicating specificity of PRO20 for PRR. In cultured human neuroblastoma cells, PRO20 blocked prorenin-induced calcium influx in a concentration- and AT(1) receptor-dependent manner. Intracerebroventricular infusion of PRO20 dose-dependently inhibited prorenin-induced hypertension in C57Bl6/J mice. Furthermore, acute intracerebroventricular infusion of PRO20 reduced blood pressure in both DOCA-salt and genetically hypertensive mice. Chronic intracerebroventricular infusion of PRO20 attenuated the development of hypertension and the increase in brain hypothalamic angiotensin II levels induced by DOCA-salt. In addition, chronic intracerebroventricular infusion of PRO20 improved autonomic function and spontaneous baroreflex sensitivity in mice treated with DOCA-salt. In summary, PRO20 binds to both mouse and human PRRs and decreases angiotensin II formation and hypertension induced by either prorenin or DOCA-salt. Our findings highlight the value of the novel PRR antagonist, PRO20, as a lead compound for a novel class of antihypertensive agents and as a research tool to establish the validity of brain PRR antagonism as a strategy for treating hypertension.


Assuntos
Anti-Hipertensivos/uso terapêutico , Hipertensão/prevenção & controle , Fragmentos de Peptídeos/uso terapêutico , Receptores de Superfície Celular/antagonistas & inibidores , Renina/uso terapêutico , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Angiotensina II/análise , Angiotensina II/fisiologia , Animais , Anti-Hipertensivos/administração & dosagem , Barorreflexo/efeitos dos fármacos , Ligação Competitiva , Pressão Sanguínea/efeitos dos fármacos , Cálcio/metabolismo , Captopril/farmacologia , Linhagem Celular Tumoral , Acetato de Desoxicorticosterona/toxicidade , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Hipertensão/induzido quimicamente , Hipertensão/tratamento farmacológico , Hipertensão/genética , Hipotálamo/química , Hipotálamo/efeitos dos fármacos , Infusões Intraventriculares , Transporte de Íons/efeitos dos fármacos , Losartan/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma , Fragmentos de Peptídeos/administração & dosagem , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores de Superfície Celular/análise , Renina/administração & dosagem , Cloreto de Sódio/toxicidade , ATPases Vacuolares Próton-Translocadoras/análise , Receptor de Pró-Renina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...