Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
bioRxiv ; 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38826452

RESUMO

Background: Small artery remodeling and endothelial dysfunction are hallmarks of hypertension. Growing evidence supports a likely causal association between cardiovascular diseases and the presence of endothelial-to-mesenchymal transition (EndMT), a cellular transdifferentiation process in which endothelial cells (ECs) partially lose their identity and acquire additional mesenchymal phenotypes. EC reprogramming represents an innovative strategy in regenerative medicine to prevent deleterious effects induced by cardiovascular diseases. Methods: Using a partial reprogramming of ECs, via overexpression of Oct-3/4, Sox-2, and Klf-4 (OSK) transcription factors, we aimed to bring ECs back to a youthful phenotype in hypertensive mice. Primary ECs were infected with lentiviral vectors (LV) containing the specific EC marker cadherin 5 (Cdh5) and the fluorescent reporter enhanced green fluorescence protein (EGFP) with empty vector (LVCO) or with OSK (LV-OSK). Confocal microscopy and western blotting analysis were used to confirm the OSK overexpression. Cellular migration, senescence, and apoptosis were evaluated. Human aortic ECs (HAoECs) from male and female normotensive and hypertensive patients were analyzed after OSK or control treatments for their endothelial nitric oxide synthase (eNOS) levels, nitric oxide (NO), and genetic profile. Male and female normotensive (BPN/3J) and hypertensive (BPH/2J) mice were treated with an intravenous (i.v.) injection of LVCO or LV-OSK and evaluated 10 days post-infection. The blood pressure, cardiac function, vascular reactivity of small arteries, in vivo EGFP signal and EndMT inhibition were analyzed. Results: OSK overexpression induced partial EC reprogramming in vitro , and these cells showed endothelial progenitor cell (EPC)-like features with lower migratory capability. OSK treatment of hypertensive BPH/2J mice normalized blood pressure and resistance arteries hypercontractility, via the attenuation of EndMT and elastin breaks. EGFP signal was detected in vivo in the prefrontal cortex of both BPN/3J and BPH/2J-treated mice, but OSK induced angiogenesis only in male BPN/3J mice. OSK-treated human ECs from hypertensive patients showed high eNOS activation and NO production, with low ROS formation. Single-cell RNA analysis showed that OSK alleviated EC senescence and EndMT, restoring their phenotypes in human ECs from hypertensive patients. Conclusion: Overall, these data indicate that OSK treatment and EC reprogramming can decrease blood pressure and reverse hypertension-induced vascular damage.

2.
Hypertension ; 80(6): 1258-1273, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37035922

RESUMO

BACKGROUND: Heart failure (HF) is a debilitating disease affecting >64 million people worldwide. In addition to impaired cardiovascular performance and associated systemic complications, most patients with HF suffer from depression and substantial cognitive decline. Although neuroinflammation and brain hypoperfusion occur in humans and rodents with HF, the underlying neuronal substrates, mechanisms, and their relative contribution to cognitive deficits in HF remains unknown. METHODS: To address this critical gap in our knowledge, we used a well-established HF rat model that mimics clinical outcomes observed in the human population, along with a multidisciplinary approach combining behavioral, electrophysiological, neuroanatomical, molecular and systemic physiological approaches. RESULTS: Our studies support neuroinflammation, hypoperfusion/hypoxia, and neuronal deficits in the hippocampus of HF rats, which correlated with the progression and severity of the disease. An increased expression of AT1aRs (Ang II [angiotensin II] receptor type 1a) in hippocampal microglia preceded the onset of neuroinflammation. Importantly, blockade of AT1Rs with a clinically used therapeutic drug (Losartan), and delivered in a clinically relevant manner, efficiently reversed neuroinflammatory end points (but not hypoxia ones), resulting in turn in improved cognitive performance in HF rats. Finally, we show than circulating Ang II can leak and access the hippocampal parenchyma in HF rats, constituting a possible source of Ang II initiating the neuroinflammatory signaling cascade in HF. CONCLUSIONS: In this study, we identified a neuronal substrate (hippocampus), a mechanism (Ang II-driven neuroinflammation) and a potential neuroprotective therapeutic target (AT1aRs) for the treatment of cognitive deficits in HF.


Assuntos
Disfunção Cognitiva , Insuficiência Cardíaca , Ratos , Humanos , Animais , Angiotensina II/farmacologia , Doenças Neuroinflamatórias , Insuficiência Cardíaca/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Hipocampo
3.
Int J Mol Sci ; 24(3)2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36768443

RESUMO

Stroke is a life-threatening condition in which accurate diagnoses and timely treatment are critical for successful neurological recovery. The current acute treatment strategies, particularly non-invasive interventions, are limited, thus urging the need for novel therapeutical targets. Arginine vasopressin (AVP) receptor antagonists are emerging as potential targets to treat edema formation and subsequent elevation in intracranial pressure, both significant causes of mortality in acute stroke. Here, we summarize the current knowledge on the mechanisms leading to AVP hyperexcretion in acute stroke and the subsequent secondary neuropathological responses. Furthermore, we discuss the work supporting the predictive value of measuring copeptin, a surrogate marker of AVP in stroke patients, followed by a review of the experimental evidence suggesting AVP receptor antagonists in stroke therapy. As we highlight throughout the narrative, critical gaps in the literature exist and indicate the need for further research to understand better AVP mechanisms in stroke. Likewise, there are advantages and limitations in using copeptin as a prognostic tool, and the translation of findings from experimental animal models to clinical settings has its challenges. Still, monitoring AVP levels and using AVP receptor antagonists as an add-on therapeutic intervention are potential promises in clinical applications to alleviate stroke neurological consequences.


Assuntos
Doenças do Sistema Nervoso , Acidente Vascular Cerebral , Animais , Arginina Vasopressina/uso terapêutico , Vasopressinas , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Antagonistas dos Receptores de Hormônios Antidiuréticos/uso terapêutico , Arginina , Glicopeptídeos/uso terapêutico
5.
J Nutr Biochem ; 77: 108318, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31923755

RESUMO

Hypertension is associated with gut dysbiosis and dysregulation of the gut-brain axis. Previous work has shown that probiotic treatments exert beneficial cardiovascular effects in humans and animal models of hypertension. Coupled with the evidence of elevated sympathetic outflow and chronic inflammation in hypertension, we hypothesized that both peripherally and centrally mediated mechanisms underlie the antihypertensive effects of kefir, a probiotic obtained from the fermentation of milk by kefir grains. Eight-week-old spontaneously hypertensive rats (SHRs) were treated by oral gavage with either vehicle or kefir (0.3 ml/100 g/day; 9 weeks; SHR-Kefir), and age-matched with vehicle-treated Wistar Kyoto rats (WKY). Long-term kefir treatment attenuated mean arterial pressure elevations in SHR-Kefir relative to vehicle-treated SHRs. Peripherally, SHRs exhibited differences in the wall of the jejunum (fewer Paneth cells per crypt of Lieberkϋhn and increased tunica muscularis thickness) and higher serum lipopolysaccharide levels compared to WKY, alterations which were reversed in SHR-Kefir. Centrally, kefir treatment reduced IL-6 and TNF-α protein densities, and abolished the microglial activation observed in the hypothalamic paraventricular nucleus and rostral ventrolateral medulla of SHRs. Taken together, our findings indicate that the antihypertensive effects of long-term kefir treatment occur, at least in part, through improved structural and functional integrity of the intestinal wall and protection against neuroinflammation within cardioregulatory nuclei.


Assuntos
Hipertensão/terapia , Kefir , Probióticos/uso terapêutico , Animais , Pressão Sanguínea , Fermentação , Microbioma Gastrointestinal , Células Caliciformes/metabolismo , Hipertensão/microbiologia , Inflamação , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Lipopolissacarídeos/metabolismo , Masculino , Celulas de Paneth/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Fator de Necrose Tumoral alfa/metabolismo
6.
Pharmacol Res ; 144: 279-291, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31039397

RESUMO

Overstimulation of the pro-inflammatory pathways within brain areas responsible for sympathetic outflow is well evidenced as a primary contributing factor to the establishment and maintenance of neurogenic hypertension. However, the precise mechanisms and stimuli responsible for promoting a pro-inflammatory state are not fully elucidated. Recent work has unveiled novel compounds derived from omega-3 polyunsaturated fatty acids (ω-3 PUFAs), termed specialized pro-resolving mediators (SPMs), which actively regulate the resolution of inflammation. Failure or dysregulation of the resolution process has been linked to a variety of chronic inflammatory and neurodegenerative diseases. Given the pathologic role of neuroinflammation in the hypertensive state, SPMs and their associated pathways may provide a link between hypertension and the long-standing association of dietary ω-3 PUFAs with cardioprotection. Herein, we review recent progress in understanding the RAS-driven pathophysiology of neurogenic hypertension, particularly in regards to the chronic low-grade neuroinflammatory response. In addition, we examine the potential for an impaired resolution of inflammation process in the context of hypertension.


Assuntos
Ácidos Graxos Ômega-3/imunologia , Hipertensão/imunologia , Inflamação/imunologia , Sistema Renina-Angiotensina , Animais , Humanos , Hipertensão/complicações , Hipertensão/fisiopatologia , Inflamação/complicações , Inflamação/fisiopatologia , Doenças do Sistema Nervoso/complicações , Doenças do Sistema Nervoso/imunologia , Doenças do Sistema Nervoso/fisiopatologia
7.
Br J Pharmacol ; 176(12): 1864-1879, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-29981161

RESUMO

The immune system plays a prominent role in the initiation and maintenance of hypertension. The innate immune system, via toll-like receptors (TLRs), identifies distinct signatures of invading microbes and damage-associated molecular patterns and triggers a chain of downstream signalling cascades, leading to secretion of pro-inflammatory cytokines and shaping the adaptive immune response. Over the past decade, a dysfunctional TLR-mediated response, particularly via TLR4, has been suggested to support a chronic inflammatory state in hypertension, inducing deleterious local and systemic effects in host cells and tissues and contributing to disease progression. While the underlying mechanisms triggering TLR4 need further research, evidence suggests that sustained elevations in BP disrupt homeostasis, releasing endogenous TLR4 ligands in hypertension. In this review, we discuss the emerging role of TLR4 in the pathogenesis of hypertension and whether targeting this receptor and its signalling pathways could offer a therapeutic strategy for management of this multifaceted disease. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.


Assuntos
Anti-Hipertensivos/farmacologia , Hipertensão/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Humanos , Hipertensão/imunologia , Ligantes , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia
8.
J Sex Med ; 15(9): 1235-1245, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30145096

RESUMO

INTRODUCTION: While increased toll-like receptor (TLR)4 activity may contribute to the pathophysiology of vascular diseases, the molecular mechanisms disrupted by this receptor in the vasculature are still poorly understood. Additionally, it is unknown if TLR4 mediates erectile dysfunction (ED) during diabetes. AIM: To investigate whether pharmacological blockade of TLR4 affects erectile function in a murine model of diabetes. METHODS: Sprague Dawley rats (Charles River Laboratory, Wilmington, MA, USA) received a single streptozotocin injection (65 mg/kg, 28 days) and were treated with an anti-TLR4 antibody (1 µg/d, intraperitoneally) for the last 14 days of the treatment. Additionally, cavernosal strips were acutely incubated for 30 minutes with CLI-095 (10-5 mol/L), a TLR4 inhibitor. Functional studies, Western blotting, erectile function, immunohistochemistry, and biochemical analyses were performed. MAIN OUTCOME MEASURES: Oxidative stress, cyclic guanosine monophosphate (cGMP) levels, and functional studies were evaluated in treated and nontreated cavernosal strips from control and diabetic animals. Additionally, in vivo erectile function was assessed. RESULTS: Enhanced TLR4 expression was observed in corpus cavernosum from diabetic rats compared with control animals. Long-term blockade of TLR4 slightly improved diabetes-induced ED in rats due to attenuation of oxidative stress and increased cGMP levels in penile tissue, which ameliorated cavernosal relaxation. Functional experiments revealed that acute or chronic inhibition of TLR4 decreased hypercontractility in response to phenylephrine and improved nitrergic relaxation in corpus cavernosum from diabetic rats. CLINICAL IMPLICATIONS: TLR4 blockade may be a novel therapeutic strategy to assist in ED management. STRENGTHS & LIMITATIONS: The strength of this article stems from the fact that we showed that TLR4 blockade partly improves erectile function in vivo in diabetic rats. Its limitations mainly include that messenger RNA analysis for the nitric oxide/cGMP pathway were not performed. CONCLUSION: In summary, TLR4 participates in the mechanisms of diabetes-associated ED and blockade of this receptor positively affects penile vascular function. Nunes KP, de Oliveira AA, Szasz T, et al. Blockade of Toll-Like Receptor 4 Attenuates Erectile Dysfunction in Diabetic Rats. J Sex Med 2018;15:1235-1245.


Assuntos
Diabetes Mellitus Experimental/complicações , Disfunção Erétil/fisiopatologia , Ereção Peniana/efeitos dos fármacos , Sulfonamidas/farmacologia , Receptor 4 Toll-Like/metabolismo , Animais , Modelos Animais de Doenças , Disfunção Erétil/complicações , Masculino , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Estreptozocina/administração & dosagem , Receptor 4 Toll-Like/antagonistas & inibidores
9.
Nutrition ; 51-52: 66-72, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29605766

RESUMO

OBJECTIVES: We aimed to evaluate whether long-term treatment with the soluble non-bacterial fraction of kefir affects mean arterial pressure (MAP) and cardiac hypertrophy through the modulation of baroreflex sensitivity, ACE activity, and the inflammatory-to-anti-inflammatory cytokine ratio in spontaneously hypertensive rats (SHRs). METHODS: SHRs were treated with the soluble non-bacterial kefir fraction (SHR-kefir) or with kefir vehicle (SHR-soluble fraction of milk). Normotensive control Wistar Kyoto animals received the soluble fraction of milk. All treatments were administered by gavage (0.3 mL/100g/body weight), once daily for eight weeks. At the end, after basal MAP and Heart Rate (HT) measurement, barorreflex sensitivity was evaluated through in bolus administrations of sodium nitroprusside and phenylephrine (AP50 [arterial pressure 50%], the lower plateau, and HR range were measured). ACE activity and cytokines (TNF-α and IL-10) were evaluated by ELISA. Cardiac hypertrophy was analysed morphometrically. RESULTS: Compared to SHR control, SHR-kefir exhibited a significant decrease in both MAP (SHR: 184 ± 5; SHR-Kefir: 142 ± 8 mmHg), and HR (SHR: 360 ± 10; SHR-kefir: 310 ± 14 bpm). The non-bacterial fraction of kefir also reduced cardiac hypertrophy, TNF-α-to-IL10 ratio, and ACE activity in SHRs. SHR-kefir baroreflex sensitivity, resulted in a partial but significant recovery of baroreflex gain, as demonstrated by improvements in AP50, the lower plateau, and HR range. CONCLUSION: In summary, our results indicate that long-term administration of the non-bacterial fraction of kefir promotes a significant decrease in both MAP and HR, by improving baroreflex, and reduces cardiac hypertrophy in SHRs, likely via ACE inhibition, and reduction of the TNF-α-to-IL10 ratio.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/sangue , Barorreflexo/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/dietoterapia , Hipertensão/dietoterapia , Kefir , Animais , Cardiomegalia/sangue , Modelos Animais de Doenças , Frequência Cardíaca , Hipertensão/sangue , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
10.
Pharmacol Res ; 120: 88-96, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28330785

RESUMO

Hypertension is a multifactorial disease. Although a number of different underlying mechanisms have been learned from the various experimental models of the disease, hypertension still poses challenges for treatment. Angiotensin II plays an unquestionable role in blood pressure regulation acting through central and peripheral mechanisms. During hypertension, dysregulation of the Renin-Angiotensin System is associated with increased expression of pro-inflammatory cytokines and reactive oxygen species causing kidney damage, endothelial dysfunction, and increase in sympathetic activity, among other damages, eventually leading to decline in organ function. Recent studies have shown that these effects involve both the innate and the adaptive immune response. The contribution of adaptive immune responses involving different lymphocyte populations in various models of hypertension has been extensively studied. However, the involvement of the innate immunity mediating inflammation in hypertension is still not well understood. The innate and adaptive immune systems intimately interact with one another and are essential to an effectively functioning of the immune response; hence, the importance of a better understanding of the underlying mechanisms mediating innate immune system during hypertension. In this review, we aim to discuss mechanisms linking Angiotensin II and the innate immune system, in the pathogenesis of hypertension. The newest research investigating Angiotensin II triggering toll like receptor 4 activation in the kidney, vasculature and central nervous system contributing to hypertension will be discussed. Understanding the role of the innate immune system in the development of hypertension may bring to light new insights necessary to improve hypertension management.


Assuntos
Angiotensina II/imunologia , Hipertensão/imunologia , Receptor 4 Toll-Like/imunologia , Imunidade Adaptativa , Animais , Encéfalo/imunologia , Encéfalo/patologia , Humanos , Hipertensão/complicações , Hipertensão/patologia , Imunidade Inata , Inflamação/complicações , Inflamação/imunologia , Inflamação/patologia , Rim/imunologia , Rim/patologia , Sistema Renina-Angiotensina , Transdução de Sinais
11.
Am J Physiol Heart Circ Physiol ; 310(3): H404-15, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26637556

RESUMO

ANG II is thought to increase sympathetic outflow by increasing oxidative stress and promoting local inflammation in the paraventricular nucleus (PVN) of the hypothalamus. However, the relative contributions of inflammation and oxidative stress to sympathetic drive remain poorly understood, and the underlying cellular and molecular targets have yet to be examined. ANG II has been shown to enhance Toll-like receptor (TLR)4-mediated signaling on microglia. Thus, in the present study, we aimed to determine whether ANG II-mediated activation of microglial TLR4 signaling is a key molecular target initiating local oxidative stress in the PVN. We found TLR4 and ANG II type 1 (AT1) receptor mRNA expression in hypothalamic microglia, providing molecular evidence for the potential interaction between these two receptors. In hypothalamic slices, ANG II induced microglial activation within the PVN (∼65% increase, P < 0.001), an effect that was blunted in the absence of functional TLR4. ANG II increased ROS production, as indicated by dihydroethidium fluorescence, within the PVN of rats and mice (P < 0.0001 in both cases), effects that were also dependent on the presence of functional TLR4. The microglial inhibitor minocycline attenuated ANG II-mediated ROS production, yet ANG II effects persisted in PVN single-minded 1-AT1a knockout mice, supporting the contribution of a non-neuronal source (likely microglia) to ANG II-driven ROS production in the PVN. Taken together, these results support functional interactions between AT1 receptors and TLR4 in mediating ANG II-dependent microglial activation and oxidative stress within the PVN. More broadly, our results support a functional interaction between the central renin-angiotensin system and innate immunity in the regulation of neurohumoral outflows from the PVN.


Assuntos
Angiotensina II/metabolismo , Microglia/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptor 4 Toll-Like/metabolismo , Angiotensina II/imunologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Antibacterianos/farmacologia , Imunidade Inata/imunologia , Inflamação , Losartan/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/imunologia , Minociclina/farmacologia , Estresse Oxidativo/genética , Estresse Oxidativo/imunologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/imunologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/imunologia , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/imunologia , Receptor 4 Toll-Like/imunologia
12.
Hypertension ; 63(3): 572-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24343120

RESUMO

Angiotensin II-mediated vascular brain inflammation emerged as a novel pathophysiological mechanism in neurogenic hypertension. However, the precise underlying mechanisms and functional consequences in relation to blood-brain barrier (BBB) integrity and central angiotensin II actions mediating neurohumoral activation in hypertension are poorly understood. Here, we aimed to determine whether BBB permeability within critical hypothalamic and brain stem regions involved in neurohumoral regulation was altered during hypertension. Using digital imaging quantification after intravascularly injected fluorescent dyes and immunohistochemistry, we found increased BBB permeability, along with altered key BBB protein constituents, in spontaneously hypertensive rats within the hypothalamic paraventricular nucleus, the nucleus of the solitary tract, and the rostral ventrolateral medulla, all critical brain regions known to contribute to neurohumoral activation during hypertension. BBB disruption, including increased permeability and downregulation of constituent proteins, was prevented in spontaneously hypertensive rats treated with the AT1 receptor antagonist losartan, but not with hydralazine, a direct vasodilator. Importantly, we found circulating angiotensin II to extravasate into these brain regions, colocalizing with neurons and microglial cells. Taken together, our studies reveal a novel angiotensin II-mediated feed-forward mechanism during hypertension, by which circulating angiotensin II evokes increased BBB permeability, facilitating in turn its access to critical brain regions known to participate in blood pressure regulation.


Assuntos
Angiotensina II/sangue , Pressão Sanguínea , Barreira Hematoencefálica/fisiopatologia , Tronco Encefálico/metabolismo , Hipertensão/metabolismo , Hipotálamo/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Tronco Encefálico/fisiopatologia , Modelos Animais de Doenças , Hipertensão/fisiopatologia , Hipotálamo/fisiopatologia , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Wistar
13.
J Comp Neurol ; 518(5): 567-85, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20034060

RESUMO

An imbalance of excitatory and inhibitory functions has been shown to contribute to numerous pathological disorders. Accumulating evidence supports the idea that a change in hypothalamic gamma-aminobutyric acid (GABA)-ergic inhibitory and glutamatergic excitatory synaptic functions contributes to exacerbated neurohumoral drive in prevalent cardiovascular disorders, including hypertension. However, the precise underlying mechanisms and neuronal substrates are still not fully elucidated. In the present study, we combined quantitative immunohistochemistry with neuronal tract tracing to determine whether plastic remodeling of afferent GABAergic and glutamatergic inputs into identified RVLM-projecting neurons of the hypothalamic paraventricular nucleus (PVN-RVLM) contributes to an imbalanced excitatory/inhibitory function in renovascular hypertensive rats (RVH). Our results indicate that both GABAergic and glutamatergic innervation densities increased in oxytocin-positive, PVN-RVLM (OT-PVN-RVLM) neurons in RVH rats. Despite this concomitant increase, time-dependent and compartment-specific differences in the reorganization of these inputs resulted in an altered balance of excitatory/inhibitory inputs in somatic and dendritic compartments. A net predominance of excitatory over inhibitory inputs was found in OT-PVN-RVLM proximal dendrites. Our results indicate that, along with previously described changes in neurotransmitter release probability and postsynaptic receptor function, remodeling of GABAergic and glutamatergic afferent inputs contributes as an underlying mechanism to the altered excitatory/inhibitory balance in the PVN of hypertensive rats.


Assuntos
Ácido Glutâmico/metabolismo , Hipertensão Renovascular/metabolismo , Bulbo/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Biomarcadores/metabolismo , Sistema Cardiovascular/fisiopatologia , Dendritos/metabolismo , Dendritos/ultraestrutura , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Glutamato Descarboxilase/metabolismo , Hipertensão Renovascular/fisiopatologia , Imuno-Histoquímica , Potenciais Pós-Sinápticos Inibidores/fisiologia , Rim/fisiopatologia , Masculino , Bulbo/citologia , Vias Neurais/citologia , Vias Neurais/metabolismo , Neurônios/citologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Ratos , Ratos Wistar , Formação Reticular/citologia , Formação Reticular/metabolismo , Transmissão Sináptica/fisiologia , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
14.
São Paulo; s.n; 2005. [126] p.
Tese em Português | LILACS | ID: lil-419529

RESUMO

O objetivo deste estudo foi avaliar os padrões hemodinâmicos de animais hipertensos renovasculares e animais hipertensos induzidos por L-NAME registrados acordados. Além disso, o sistema nervoso simpático e alterações de neurotransmissores excitatórios e inibitórios em um núcleo pré-motor do simpático em animais renovasculares; bem como, o sistema nervoso simpático e o sistema renina-angiotensina em animais hipertensos por inibição da síntese de óxido nítrico também foram avaliados. Animais renovasculares modelo Goldblatt dois rins um clipe foram estudados seis semanas pós clipagem. O padrão hemodinâmico desses animais registrados acordados demonstrou aumento da pressão arterial média com aumento de resistência periférica total e débito cardíaco normal. O bloqueio do sistema nervoso simpático produziu queda de pressão arterial exclusivamente em função da queda de resistência periférica total, demonstrando a ativação simpática que ocorre nesta fase. Em neurônios do núcleo paraventricular do hipotálamo que se projetam para a região rostroventrolateral, dois núcleos importantes no controle cardiovascular, foram encontradas alterações tanto na inervação GABAérgica, como na glutamatérgica, estando ambas aumentadas neste modelo. Considerando que na região rostroventrolateral a atividade dos neurônios pré-sinápticos são determinados por um balanço de respostas inibitórias e excitatórias, e sabendo-se que nesta região há aumento da ação glutamatérgica previamente demonstrada, pode-se inferir a possibilidade de que, no lugar apenas de uma inibição GABAérgica e ativação glutamatérgica, tenha-se um balanço positivo decorrente do aumento de ambos os neurotransmissores em função de um possível mecanismo compensatório…(au)


Assuntos
Hipertensão Renovascular , Óxido Nítrico , Hemodinâmica , Sistema Renina-Angiotensina , Sistema Nervoso Simpático
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA