Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 57(9): 2000-2002, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39260350

RESUMO

Our brain is not an immune-privileged island isolated from peripheries, but how non-neuronal brain cells interact with the peripheral system is not well understood. Wei et al. report that microglia in the hypothalamic paraventricular nucleus (PVN) with unique vasculature can detect ATP derived from hemodynamic disturbance. These microglia in the PVN regulate the response to hypertension via ATP-P2Y12-C/EBPß signaling.


Assuntos
Pressão Sanguínea , Encéfalo , Microglia , Núcleo Hipotalâmico Paraventricular , Microglia/imunologia , Microglia/fisiologia , Microglia/metabolismo , Animais , Humanos , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Pressão Sanguínea/fisiologia , Encéfalo/imunologia , Trifosfato de Adenosina/metabolismo , Transdução de Sinais , Hipertensão/imunologia , Hipertensão/fisiopatologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo
2.
Am J Physiol Heart Circ Physiol ; 317(2): H279-H289, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31150271

RESUMO

Increased sympathetic nervous system activity is a hallmark of hypertension (HTN), and it is implicated in altered immune system responses in its pathophysiology. However, the precise mechanisms of neural-immune interaction in HTN remain elusive. We have previously shown an association between elevated sympathetic drive to the bone marrow (BM) and activated BM immune cells in rodent models of HTN. Moreover, microglial-dependent neuroinflammation is also seen in rodent models of HTN. However, the cause-effect relationship between central and systemic inflammatory responses and the sympathetic drive remains unknown. These observations led us to hypothesize that increase in the femoral BM sympathetic nerve activity (fSNA) initiates a cascade of events leading to increase in blood pressure (BP). Here, we investigated the temporal relationship between the BM sympathetic drive, activation of the central and peripheral immune system, and increase in BP in the events leading to established HTN. The present study demonstrates that central infusion of angiotensin II (ANG II) induces early microglial activation in the paraventricular nucleus of hypothalamus, which preceded increase in the fSNA. In turn, activation of fSNA correlated with the timing of increased production and release of CD4+.IL17+ T cells and other proinflammatory cells into circulation and elevation in BP, whereas infiltration of CD4+ cells to the paraventricular nucleus marked establishment of ANG II HTN. This study identifies cellular and molecular mechanisms involved in neural-immune interactions in early and established stages of rodent ANG II HTN. NEW & NOTEWORTHY Early microglia activation in paraventricular nucleus precedes sympathetic activation of the bone marrow. This leads to increased bone marrow immune cells and their release into circulation and an increase in blood pressure. Infiltration of CD4+ T cells into paraventricular nucleus paraventricular nucleus marks late hypertension.


Assuntos
Pressão Sanguínea , Medula Óssea/inervação , Hipertensão/fisiopatologia , Inflamação/fisiopatologia , Neuroimunomodulação , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Angiotensina II , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Fêmur , Hipertensão/induzido quimicamente , Hipertensão/imunologia , Hipertensão/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Microglia/imunologia , Microglia/metabolismo , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Sprague-Dawley , Sistema Nervoso Simpático/imunologia , Sistema Nervoso Simpático/metabolismo , Fatores de Tempo
3.
Int J Neuropsychopharmacol ; 22(2): 137-142, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30535261

RESUMO

High dietary salt intake increases risk of stress-related neuropsychiatric disorders. Here, we explored the contribution of high dietary salt intake-induced neuroinflammation in key stress-responsive brain regions, the hypothalamic paraventricular nucleus and basolateral amygdala, in promoting exaggerated neuronal activation and coping behaviors in response to acute psychogenic stress. Mice that underwent high dietary salt intake exhibited increased active stress coping behaviors during and after an acute swim stress, and these were reduced by concurrent administration of minocycline, an inhibitor of microglial activation, without affecting body fluid hyperosmolality caused by high dietary salt intake. Moreover, minocycline attenuated high dietary salt intake-induced increases of paraventricular nucleus tumor necrosis factor-α, activated microglia (ionized calcium-binding adaptor molecule 1), and acute swim stress-induced neuronal activation (c-Fos). In the basolateral amygdala, similar effects were observed on ionized calcium-binding adaptor molecule 1+ and c-Fos+ counts, but not tumor necrosis factor-α levels. These data indicate that high dietary salt intake promotes neuroinflammation, increasing recruitment of neurons in key stress-associated brain regions and augmenting behavioral hyper-responsivity to acute psychological stress.


Assuntos
Adaptação Psicológica , Anti-Inflamatórios/farmacologia , Complexo Nuclear Basolateral da Amígdala , Encéfalo , Inflamação , Microglia , Minociclina/farmacologia , Núcleo Hipotalâmico Paraventricular , Cloreto de Sódio na Dieta/efeitos adversos , Estresse Psicológico , Adaptação Psicológica/efeitos dos fármacos , Animais , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/imunologia , Complexo Nuclear Basolateral da Amígdala/metabolismo , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Inflamação/complicações , Inflamação/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/imunologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/imunologia , Estresse Psicológico/fisiopatologia
4.
Neurochem Res ; 43(9): 1802-1813, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30030770

RESUMO

Systemic inflammation is present in obesity and emerging evidence, primarily from studies using male rodents fed high-fat diets, suggests neuroimmune signaling also is involved. We investigated early changes in neuroimmune signaling during the weight gain that follows ovariectomy in rats. Ovariectomized (OVX) rats were given standard rat chow and terminated 5 days (baseline), 4 or 8 weeks after ovariectomy. Levels of interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) in plasma and periuterine adipose were not affected by ovariectomy. In contrast, compared to baseline levels, IL-6 expression in the arcuate nucleus (ARC) and dorsal vagal complex (DVC) decreased by 4 weeks after OVX, but was not affected in the paraventricular nucleus (PVN). MCP-1 expression decreased by 4 weeks in the ARC and by 8 weeks in the PVN, but was not affected in the DVC. Increased glial fibrillary acidic protein (GFAP) expression in the PVN indicated astrocyte activation; decreased toll-like receptor 4 (TLR4) expression in the ARC, but not other regions, suggested early effects on innate immune factors. Importantly, in reproductively intact rats, IL-6 and MCP-1 levels in plasma, periuterine adipose, and brain regions were not affected after 8 weeks. Unlike OVX rats, GFAP expression in the DVC of intact rats was decreased at 8 weeks, and TLR4 expression in the ARC was increased at 8 weeks. Taken together, these dynamic and selective changes in neuroimmune factors co-incident with post-ovariectomy weight gain provide insight into the role of neuroimmune signaling in obesity, particularly in females.


Assuntos
Encéfalo/imunologia , Obesidade/etiologia , Ovariectomia/efeitos adversos , Núcleo Hipotalâmico Paraventricular/metabolismo , Aumento de Peso/imunologia , Animais , Encéfalo/metabolismo , Estradiol/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/imunologia , Obesidade/imunologia , Núcleo Hipotalâmico Paraventricular/imunologia , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley
5.
Circ Res ; 117(2): 178-91, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25963715

RESUMO

RATIONALE: Microglial activation in autonomic brain regions is a hallmark of neuroinflammation in neurogenic hypertension. Despite evidence that an impaired sympathetic nerve activity supplying the bone marrow (BM) increases inflammatory cells and decreases angiogenic cells, little is known about the reciprocal impact of BM-derived inflammatory cells on neuroinflammation in hypertension. OBJECTIVE: To test the hypothesis that proinflammatory BM cells from hypertensive animals contribute to neuroinflammation and hypertension via a brain-BM interaction. METHODS AND RESULTS: After BM ablation in spontaneously hypertensive rats, and reconstitution with normotensive Wistar Kyoto rat BM, the resultant chimeric spontaneously hypertensive rats displayed significant reduction in mean arterial pressure associated with attenuation of both central and peripheral inflammation. In contrast, an elevated mean arterial pressure along with increased central and peripheral inflammation was observed in chimeric Wistar-Kyoto rats reconstituted with spontaneously hypertensive rat BM. Oral treatment with minocycline, an inhibitor of microglial activation, attenuated hypertension in both the spontaneously hypertensive rats and the chronic angiotensin II-infused rats. This was accompanied by decreased sympathetic drive and inflammation. Furthermore, in chronic angiotensin II-infused rats, minocycline prevented extravasation of BM-derived cells to the hypothalamic paraventricular nucleus, presumably via a mechanism of decreased C-C chemokine ligand 2 levels in the cerebrospinal fluid. CONCLUSIONS: The BM contributes to hypertension by increasing peripheral inflammatory cells and their extravasation into the brain. Minocycline is an effective therapy to modify neurogenic components of hypertension. These observations support the hypothesis that BM-derived cells are involved in neuroinflammation, and targeting them may be an innovative strategy for neurogenic resistant hypertension therapy.


Assuntos
Células da Medula Óssea/fisiologia , Hipertensão/etiologia , Microglia/fisiologia , Inflamação Neurogênica/complicações , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Angiotensina II , Animais , Barorreflexo/fisiologia , Transplante de Medula Óssea , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Feminino , Hipertensão/fisiopatologia , Hipertensão/prevenção & controle , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Masculino , Microglia/efeitos dos fármacos , Minociclina/uso terapêutico , Norepinefrina/sangue , Núcleo Hipotalâmico Paraventricular/imunologia , Quimera por Radiação , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Sistema Nervoso Simpático/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia
6.
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
7.
Am J Physiol Heart Circ Physiol ; 309(7): H1115-22, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26254332

RESUMO

Exercise training (ExT) is recommended to treat hypertension along with pharmaceutical antihypertensive therapies. Effects of ExT in hypothalamic content of high mobility box 1 (HMGB1) and microglial activation remain unknown. We examined whether ExT would decrease autonomic and cardiovascular abnormalities in spontaneously hypertensive rats (SHR), and whether these effects were associated with decreased HMGB1 content, microglial activation, and inflammation in the hypothalamic paraventricular nucleus (PVN). Normotensive Wistar-Kyoto (WKY) rats and SHR underwent moderate-intensity ExT for 2 wk. After ExT, cardiovascular (heart rate and arterial pressure) and autonomic parameters (arterial pressure and heart rate variability, peripheral sympathetic activity, cardiac vagal activity, and baroreflex function) were measured in conscious and freely-moving rats through chronic arterial and venous catheterization. Cerebrospinal fluid, plasma, and brain were collected for molecular and immunohistochemistry analyses of the PVN. In addition to reduced heart rate variability, decreased vagal cardiac activity and increased mean arterial pressure, heart rate, arterial pressure variability, cardiac, and vasomotor sympathetic activity, SHR had higher HMGB1 protein expression, IκB-α phosphorylation, TNF-α and IL-6 protein expression, and microglia activation in the PVN. These changes were accompanied by higher plasma and cerebrospinal fluid levels of HMGB1. The ExT + SHR group had decreased expression of HMGB1, CXCR4, SDF-1, and phosphorylation of p42/44 and IκB-α. ExT reduced microglial activation and proinflammatory cytokines content in the PVN, and improved autonomic control as well. Data suggest that training-induced downregulation of activated HMGB1/CXCR4/microglia/proinflammatory cytokines axis in the PVN of SHR is a prompt neural adaptation to counterbalance the deleterious effects of inflammation on autonomic control.


Assuntos
Citocinas/metabolismo , Proteína HMGB1/metabolismo , Microglia/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Condicionamento Físico Animal , Sistema Nervoso Simpático/fisiopatologia , Animais , Pressão Arterial , Sistema Nervoso Autônomo/fisiologia , Sistema Nervoso Autônomo/fisiopatologia , Barorreflexo/fisiologia , Citocinas/imunologia , Frequência Cardíaca/fisiologia , Proteínas I-kappa B/metabolismo , Inflamação , Interleucina-6/imunologia , Interleucina-6/metabolismo , Microglia/fisiologia , Inibidor de NF-kappaB alfa , Núcleo Hipotalâmico Paraventricular/imunologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores CXCR4/imunologia , Receptores CXCR4/metabolismo , Transdução de Sinais , Sistema Nervoso Simpático/fisiologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Nervo Vago/fisiologia , Nervo Vago/fisiopatologia
8.
Toxicol Appl Pharmacol ; 284(3): 315-22, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25759242

RESUMO

We hypothesized that chronic inhibition of NF-κB activity in the hypothalamic paraventricular nucleus (PVN) delays the progression of hypertension and attenuates cardiac hypertrophy by up-regulating anti-inflammatory cytokines, reducing pro-inflammatory cytokines (PICs), attenuating nuclear factor-κB (NF-κB) p65 and NAD(P)H oxidase in the PVN of young spontaneously hypertensive rats (SHR). Young normotensive Wistar-Kyoto (WKY) and SHR rats received bilateral PVN infusions with NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) or vehicle for 4 weeks. SHR rats had higher mean arterial pressure and cardiac hypertrophy as indicated by increased whole heart weight/body weight ratio, whole heart weight/tibia length ratio, left ventricular weight/tibia length ratio, cardiomyocyte diameters of the left cardiac ventricle, and mRNA expressions of cardiac atrial natriuretic peptide (ANP) and beta-myosin heavy chain (ß-MHC). These SHR rats had higher PVN levels of proinflammatory cytokines (PICs), reactive oxygen species (ROS), the chemokine monocyte chemoattractant protein-1 (MCP-1), NAD(P)H oxidase activity, mRNA expression of NOX-2 and NOX-4, and lower PVN IL-10, and higher plasma levels of PICs and NE, and lower plasma IL-10. PVN infusion of NF-κB inhibitor PDTC attenuated all these changes. These findings suggest that NF-κB activation in the PVN increases sympathoexcitation and hypertensive response, which are associated with the increases of PICs and oxidative stress in the PVN; PVN inhibition of NF-κB activity attenuates PICs and oxidative stress in the PVN, thereby attenuates hypertension and cardiac hypertrophy.


Assuntos
Cardiomegalia/prevenção & controle , Citocinas/metabolismo , Hipertensão/tratamento farmacológico , Mediadores da Inflamação/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia , Fator de Transcrição RelA/antagonistas & inibidores , Animais , Pressão Arterial/efeitos dos fármacos , Biomarcadores/metabolismo , Cardiomegalia/etiologia , Cardiomegalia/imunologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Hipertensão/complicações , Hipertensão/imunologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Fatores de Tempo , Fator de Transcrição RelA/metabolismo
9.
Toxicol Appl Pharmacol ; 279(2): 141-9, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24937322

RESUMO

The hypothalamic paraventricular nucleus (PVN) and rostral ventrolateral medulla (RVLM) play a critical role in the generation and maintenance of sympathetic nerve activity. The renin-angiotensin system (RAS) in the brain is involved in the pathogenesis of hypertension. This study was designed to determine whether inhibition of the angiotensin-converting enzyme (ACE) in the PVN modulates cytokines and attenuates oxidative stress (ROS) in the RVLM, and decreases the blood pressure and sympathetic activity in renovascular hypertensive rats. Renovascular hypertension was induced in male Sprague-Dawley rats by the two-kidney one-clip (2K1C) method. Renovascular hypertensive rats received bilateral PVN infusion with ACE inhibitor lisinopril (LSP, 10µg/h) or vehicle via osmotic minipump for 4weeks. Mean arterial pressure (MAP), renal sympathetic nerve activity (RSNA), and plasma proinflammatory cytokines (PICs) were significantly increased in renovascular hypertensive rats. The renovascular hypertensive rats also had higher levels of ACE in the PVN, and lower level of interleukin-10 (IL-10) in the RVLM. In addition, the levels of PICs, the chemokine MCP-1, the subunit of NAD(P)H oxidase (gp91(phox)) and ROS in the RVLM were increased in hypertensive rats. PVN treatment with LSP attenuated those changes occurring in renovascular hypertensive rats. Our findings suggest that the beneficial effects of ACE inhibition in the PVN in renovascular hypertension are partly due to modulation cytokines and attenuation oxidative stress in the RVLM.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Anti-Hipertensivos/administração & dosagem , Citocinas/metabolismo , Hipertensão Renovascular/tratamento farmacológico , Mediadores da Inflamação/metabolismo , Lisinopril/administração & dosagem , Bulbo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Animais , Anti-Inflamatórios/administração & dosagem , Antioxidantes/administração & dosagem , Pressão Arterial/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Citocinas/sangue , Modelos Animais de Doenças , Hipertensão Renovascular/imunologia , Hipertensão Renovascular/metabolismo , Hipertensão Renovascular/fisiopatologia , Mediadores da Inflamação/sangue , Infusões Parenterais , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Masculino , Bulbo/imunologia , Bulbo/metabolismo , Bulbo/fisiopatologia , Glicoproteínas de Membrana/metabolismo , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Peptidil Dipeptidase A/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia , Fatores de Tempo
10.
Brain Behav Immun ; 41: 32-43, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24975592

RESUMO

The early life period is one of significant vulnerability to programming effects from the environment. Given the sensitivity of microglial cells to early life programming and to adult diet, we hypothesized overfeeding during the neonatal period would acutely alter microglial profiles within the developing brain, predisposing the individual to a lasting central pro-inflammatory profile that contributes to overactive immune responses long-term. We tested this idea by manipulating litter sizes in which Wistar rat pups were raised, so the pups were suckled in litters of 4 (neonatally overfed) or 12 (control). This manipulation induces obesity and susceptibility to lipopolysaccharide (LPS) long-term. We then examined microglial and central pro-inflammatory profiles during development and in adulthood as well as susceptibility to neuroimmune challenge with LPS. Neonatally overfed rats have evidence of microgliosis in the paraventricular nucleus of the hypothalamus (PVN) as early as postnatal day 14. They also show changes in hypothalamic gene expression at this time, with suppressed hypothalamic interleukin 1ß mRNA. These effects persist into adulthood, with basal PVN microgliosis and increased hypothalamic toll-like receptor 4, nuclear factor κB, and interleukin 6 gene expression. These neonatally overfed rats also have dramatically exacerbated microglial activation in the PVN 24h after an adult LPS challenge, coupled with changes in inflammatory gene expression. Thus, it appears neonatal overfeeding sensitizes PVN microglia, contributing to a basal pro-inflammatory profile and an altered response to a neuroimmune challenge throughout life. It remains to be seen if these effects can be reversed with early interventions.


Assuntos
Transtornos da Nutrição do Lactente/imunologia , Sobrepeso/imunologia , Núcleo Hipotalâmico Paraventricular/imunologia , Animais , Animais Recém-Nascidos , Citocinas/biossíntese , Citocinas/genética , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transtornos da Nutrição do Lactente/genética , Transtornos da Nutrição do Lactente/patologia , Recém-Nascido , Inflamação/etiologia , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/toxicidade , Tamanho da Ninhada de Vivíparos , Masculino , NF-kappa B/biossíntese , NF-kappa B/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neuroimunomodulação/fisiologia , Sobrepeso/etiologia , Sobrepeso/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Gravidez , Ratos , Ratos Wistar , Método Simples-Cego , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/genética , Aumento de Peso
11.
Brain Behav Evol ; 79(2): 84-97, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22067598

RESUMO

The neuropeptide arginine vasotocin (AVT: the avian homolog of vasopressin) has numerous functional roles including mediating social behaviors, coregulating the adrenocortical stress response and maintaining water balance. These functions of AVT make it susceptible to environmental influence, yet little is understood concerning the variation in the AVT system across habitats. In this study, AVT immunoreactivity was compared between male curve-billed thrashers, Toxostoma curvirostre, from native Sonoran Desert locations and those within the city of Phoenix, Ariz. Previous research found that urban thrashers are more responsive to territorial intrusion, secrete more corticosterone (CORT) during capture stress, and they may also have greater access to water than desert counterparts. Variation in AVT immunoreactivity was also related to levels of plasma CORT and osmolality, and with behavioral responses to a simulated territorial intrusion. Birds from these two habitats showed different AVT immunoreactive patterns in two brain regions: the paraventricular nucleus of the hypothalamus and the medial bed nucleus of the stria terminalis (BSTM), a part of the limbic system. Immunoreactive AVT within the paraventricular nucleus was associated with plasma CORT levels in urban, but not desert, birds, but no such association with osmolality was observed in birds from either habitat. The total number of BSTM AVT-immunoreactive cells was related to a decreased responsiveness to territorial intrusion. These data suggest that divergence in the AVT system between urban and desert thrashers may help explain observed differences in both the adrenocortical stress response and territorial behavior between populations. Whether differences in water availability between habitats contribute to population differences in the brain AVT system is unknown.


Assuntos
Encéfalo/imunologia , Passeriformes/imunologia , Estresse Fisiológico/imunologia , Territorialidade , Vasotocina/imunologia , Animais , Encéfalo/anatomia & histologia , Corticosterona/sangue , Comportamento de Retorno ao Território Vital/fisiologia , Masculino , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Núcleo Hipotalâmico Paraventricular/imunologia , Passeriformes/classificação , Passeriformes/fisiologia , Núcleos Septais/anatomia & histologia , Núcleos Septais/imunologia , Estresse Fisiológico/fisiologia
12.
Bull Exp Biol Med ; 151(2): 247-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22238761
13.
Behav Brain Res ; 402: 113113, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33412227

RESUMO

Early-life stress (ELS) is a high-risk factor for the development of chronic visceral pain in adulthood. Emerging evidence suggests that mast cells play a key role in the development of visceral hypersensitivity through interaction with neurons. The sensitization of corticotropin-releasing factor (CRF) neurons in the hypothalamic paraventricular nucleus (PVN) plays a pivotal role in the pathogenesis of visceral pain. However, the precise mechanism by which mast cells and CRF neurons interact in the PVN in the pathogenesis of visceral hypersensitivity remains elusive. In the present study, we used neonatal maternal separation (MS), an ELS model, and observed that neonatal MS induced visceral hypersensitivity and triggered PVN mast cell activation in adult rats, which was repressed by intra-PVN infusion of the mast cell stabilizer disodium cromoglycate (cromolyn). Wild-type (WT) mice but not mast cell-deficient KitW-sh/W-sh mice that had experienced neonatal MS exhibited chronic visceral hypersensitivity. MS was associated with an increase in the expression of proinflammatory mediators, the number of CRF+ cells and CRF protein in the PVN, which was prevented by intra-PVN infusion of cromolyn. Furthermore, we demonstrated that intra-PVN infusion of the mast degranulator compound 48/80 significantly induced mast cell activation, resulting in proinflammatory mediator release, CRF neuronal sensitization, and visceral hypersensitivity, which was suppressed by cromolyn. Overall, our findings demonstrated that neonatal MS induces the activation of PVN mast cells, which secrete numerous proinflammatory mediators that may participate in neighboring CRF neuronal activity, ultimately directly inducing visceral hypersensitivity in adulthood.


Assuntos
Hiperalgesia , Mastócitos , Privação Materna , Núcleo Hipotalâmico Paraventricular , Estresse Psicológico , Dor Visceral , Animais , Masculino , Camundongos , Ratos , Animais Recém-Nascidos , Modelos Animais de Doenças , Hiperalgesia/etiologia , Hiperalgesia/imunologia , Hiperalgesia/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos Transgênicos , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Sprague-Dawley , Dor Visceral/imunologia , Dor Visceral/metabolismo
14.
Biomed Pharmacother ; 135: 111189, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33388596

RESUMO

Microbiota has a role in the host blood pressure (BP) regulation. The immunosuppressive drug mofetil mycophenolate (MMF) ameliorates hypertension. The present study analyzes whether MMF improves dysbiosis in a genetic model of hypertension. Twenty weeks old male spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY) were randomly divided into three groups: untreated WKY, untreated SHR, and SHR treated with MMF for 5 weeks. MMF treatment restored gut bacteria from the phyla Firmicutes and Bacteroidetes, and acetate- and lactate-producing bacteria to levels similar to those found in WKY, increasing butyrate-producing bacteria. MMF increased the percentage of anaerobic bacteria in the gut. The improvement of gut dysbiosis was associated with an enhanced colonic integrity and a decreased sympathetic drive in the gut. MMF inhibited neuroinflammation in the paraventricular nuclei in the hypothalamus. MMF increased the lower regulatory T cells proportion in mesenteric lymph nodes and Th17 and Th1 infiltration in aorta, improved aortic endothelial function and reduced systolic BP. This study demonstrates for the first time that MMF reduces gut dysbiosis in SHR. This effect could be related to its capability to improve gut integrity due to reduced sympathetic drive in the gut associated to the reduced brain neuroinflammation.


Assuntos
Anti-Hipertensivos/farmacologia , Bactérias/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Colo/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Ácido Micofenólico/farmacologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/imunologia , Aorta Torácica/fisiopatologia , Bactérias/crescimento & desenvolvimento , Bactérias/metabolismo , Células Cultivadas , Colo/inervação , Citocinas/metabolismo , Modelos Animais de Doenças , Disbiose , Hipertensão/imunologia , Hipertensão/microbiologia , Hipertensão/fisiopatologia , Mediadores da Inflamação/metabolismo , Neuroimunomodulação/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
15.
Neuroimmunomodulation ; 17(5): 305-13, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20407282

RESUMO

OBJECTIVE: In the hypothalamus, corticotropin-releasing factor (CRF) plays a central role in regulating stress responses. Cytokines are important mediators of the interaction between the neuroendocrine and immune systems, and are implicated in the regulation of CRF expression. Following inflammatory challenges, interleukin (IL)-1 or IL-6 stimulates the hypothalamic-pituitary-adrenal axis. CRF promoter contains multiple nuclear factor (NF)-kappaB and Nurr1 binding sites. In the present study, we determined the ability of the signaling pathways to activate the CRF gene in the hypothalamic paraventricular nucleus following inflammatory challenge. METHODS: Cytokine-induced changes in CRF gene expression were examined in the hypothalamic system. Luciferase assay and Western blotting were performed to assess transcriptional activity and the nuclear translocation of transcriptional factors. RESULTS: IL-1beta, IL-6 and tumor necrosis factor (TNF)-alpha stimulated the nuclear expression levels of NF-kappaB, NF-kappaB-dependent Nurr1 and c-Fos proteins. Direct stimulatory effects of TNF-alpha and IL-1beta, in addition to IL-6, were found on the transcriptional activity of the CRF gene in hypothalamic 4B cells. CONCLUSION: These cytokines are involved in the regulation of CRF gene activity in hypothalamic cells.


Assuntos
Hormônio Liberador da Corticotropina/genética , Citocinas/fisiologia , Hipotálamo/metabolismo , NF-kappa B/genética , Neurônios/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Animais , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Linhagem Celular , Hormônio Liberador da Corticotropina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Interleucina-1beta/fisiologia , Interleucina-6/fisiologia , Camundongos , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Regiões Promotoras Genéticas/imunologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
16.
Neuro Endocrinol Lett ; 31(5): 609-15, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21173750

RESUMO

OBJECTIVES AND METHODS: Neuropeptides, as the main neuroendocrine system effectors, regulate notably the response to different stressors via a secretory plasticity within their respective hypothalamic neuronal populations. The aim of the present study was to explore by immunocytochemistry the occurrence and the potential expression plasticity of the novel neuropeptide EM66 in the CRH neurones of stressed rats. RESULTS: The secretogranin II (SgII)-derived peptide EM66 is strongly expressed within hypothalamic neuroendocrine areas such as the parvocellular aspect of the paraventricular nucleus (pPVN) as well as the median eminence, suggesting a probable hypophysiotropic effect of this peptide. As a first approach to investigate such a role, we evaluated by immunohistochemistry EM66 expression within the pPVN following acute immune stress induced by lipopolysaccharide (LPS) or interleukin-1ß (IL-1ß) injection in rat. This study showed that EM66 is present in the pPVN but the number of EM66 immunolabeled cells did not fluctuate in this structure following LPS peripheral injection. In line with this observation, an intracerebroventricular injection of IL-1ß did not provoke any significant variation of the number of intraparaventricular EM66 neurones. CONCLUSION: The present data revealed for the first time that EM66 expression would be insensitive to the central and peripheral cytokines within the neurosecretory hypothalamic pPVN. This result indicates that EM66 does not participate to the phenotypic plasticity of hypothalamic parvicellular neurones in response to acute inflammatory stress.


Assuntos
Plasticidade Neuronal , Neuropeptídeos/biossíntese , Núcleo Hipotalâmico Paraventricular/metabolismo , Fragmentos de Peptídeos/metabolismo , Secretogranina II/metabolismo , Estresse Fisiológico , Animais , Modelos Animais de Doenças , Imuno-Histoquímica , Injeções Intraperitoneais , Injeções Intraventriculares , Interleucina-1beta , Lipopolissacarídeos , Masculino , Núcleo Hipotalâmico Paraventricular/imunologia , Fragmentos de Peptídeos/imunologia , Ratos , Ratos Wistar , Secretogranina II/química , Secretogranina II/imunologia , Estresse Fisiológico/imunologia
17.
Brain Res ; 1712: 93-100, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30731078

RESUMO

The ovarian hormone 17ß-estradiol is known to regulate the release, expression and immunoreactivity of arginine-vasopressin (AVP) in the supraoptic and paraventricular hypothalamic nuclei of rodents. Previous studies have shown that estrogen receptor α is involved in the effects of chronic estradiol administration on arginine-vasopressin immunoreactivity in the female rat hypothalamus. In this study we have examined the effect of an acute administration of estradiol or specific agonists for estrogen receptors α, ß and G protein-coupled estrogen receptor 1 on the immunoreactivity of arginine-vasopressin in the hypothalamus of adult ovariectomized female rats. Acute estradiol administration resulted in a significant decrease in the number of arginine-vasopressin immunoreactive neurons in the supraoptic and paraventricular nuclei after 24 h. The effects of the specific estrogen receptors agonists suggest that the action of estradiol on arginine-vasopressin immunoreactivity is mediated in the supraoptic nucleus by G protein-coupled estrogen receptor 1 and in the paraventricular nucleus by both estrogen receptor ß and G protein-coupled estrogen receptor 1. Thus, in contrast to previous studies on the effect of chronic estrogenic treatments, the present findings suggest that estrogen receptor ß and G protein-coupled estrogen receptor 1 mediate the acute effects of estradiol on arginine-vasopressin immunoreactivity in the hypothalamus of ovariectomized rats.


Assuntos
Arginina Vasopressina/metabolismo , Receptor beta de Estrogênio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Núcleo Supraóptico/metabolismo , Animais , Arginina Vasopressina/imunologia , Estradiol/farmacologia , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/imunologia , Feminino , Hipotálamo/imunologia , Hipotálamo/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , Ovariectomia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/imunologia , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/imunologia , Núcleo Supraóptico/efeitos dos fármacos , Núcleo Supraóptico/imunologia
18.
Brain Behav Immun ; 22(4): 573-89, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18191534

RESUMO

Activation of the inflammatory immune response may provoke neuroendocrine and central neurochemical effects that are reminiscent of those elicited by traditional stressors, and when administered concurrently may have synergistic effects. The present investigation assessed whether a psychosocial stressor, comprising social disruption, would augment the effects of lipopolysaccharide in mice. It was indeed observed that the social disruption engendered by a period of 2-4 weeks of social isolation (but not 1-7 days of this treatment) followed by regrouping, enhanced the effects of lipopolysaccharide (LPS: 10mug) in the provocation of sickness behavior, as well as plasma corticosterone, IL-6, TNF-alpha and IL-10 levels. Similar effects were not apparent with respect to IL-1beta, IL-4, or IFN-gamma. Synergy between LPS and other stressors (restraint, tail pinch, and loud noise) was not apparent with respect to sickness or plasma corticosterone, provisionally suggesting that social stressors, such as regrouping, may be more powerful or may engage unique neural or neuroendocrine circuits that favour synergistic outcomes. Within the CNS, the LPS and the regrouping stressor synergistically enhanced NE utilization within the prefrontal cortex, and additively influenced hippocampal NE utilization. In contrast to the effects on circulating cytokines, the LPS-induced elevation of IL-1beta, IL-6 and TNF-alpha mRNA expression in the hippocampus, PFC and nucleus tractus solitarius was diminished in animals that had experienced the regrouping stressor. In view of the combined actions of LPS challenge and a social stressor, these data are interpreted as suggesting that models of depression based on immune activation ought to consider the stressor backdrop upon which immune challenges are imposed.


Assuntos
Comportamento Animal , Encéfalo/imunologia , Endotoxemia/imunologia , Endotoxemia/psicologia , Estresse Psicológico/imunologia , Animais , Encéfalo/metabolismo , Corticosterona/sangue , Endotoxemia/induzido quimicamente , Hipocampo/imunologia , Hipocampo/metabolismo , Interleucina-1beta/sangue , Interleucina-1beta/genética , Interleucina-6/sangue , Interleucina-6/genética , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Norepinefrina/metabolismo , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Córtex Pré-Frontal/imunologia , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serotonina/metabolismo , Papel do Doente , Isolamento Social , Núcleo Solitário/imunologia , Núcleo Solitário/metabolismo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética
19.
Brain Res ; 1130(1): 130-45, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17169348

RESUMO

Immune-responsive neurons in the brainstem, primarily in the nucleus of the solitary tract (NTS) and ventrolateral medulla (VLM), contribute to a significant drive on forebrain nuclei responsible for brain-mediated host defense responses. The current study investigated the relative contribution of brainstem-derived ascending pathways to forebrain immune-responsive nuclei in the rat by means of retrograde tract tracing and c-Fos immunohistochemistry. Fluorogold was iontophoresed into the bed nucleus of stria terminalis (BST), central nucleus of the amygdala (CEA), paraventricular nucleus of the hypothalamus (PVN), and the pontine lateral parabrachial nucleus (PBL; an important component of ascending viscerosensensory pathways) followed 2 weeks later by intraperitoneal injection of lipopolysaccharide (LPS, 0.1 mg/kg) or saline. The NTS and VLM provide immune-responsive input to all four regions, via direct, predominantly catecholaminergic, projections to the PVN, the lateral BST, and the CEA, and mostly non-catecholaminergic projections to the PBL. The PBL provides a major LPS-activated input to the BST and CEA. The pattern of LPS-activated catecholaminergic projections from the VLM and NTS to the forebrain is characterized by a strong predominance of VLM input to the PVN, whereas the NTS provides a greater contribution to the BST. These findings indicate that direct and indirect pathways originate in the caudal brainstem that propagate immune-related information from the periphery with multiple levels of processing en route to the forebrain nuclei, which may allow for integration of brain responses to infection.


Assuntos
Tonsila do Cerebelo/citologia , Bulbo/citologia , Vias Neurais/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleos Septais/citologia , Tonsila do Cerebelo/imunologia , Tonsila do Cerebelo/metabolismo , Animais , Corantes Fluorescentes/metabolismo , Imuno-Histoquímica , Lipopolissacarídeos/imunologia , Masculino , Bulbo/imunologia , Bulbo/metabolismo , Vias Neurais/imunologia , Vias Neurais/metabolismo , Neuroimunomodulação/imunologia , Neuroimunomodulação/fisiologia , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Núcleos Septais/imunologia , Núcleos Septais/metabolismo , Fibras Aferentes Viscerais/citologia , Fibras Aferentes Viscerais/imunologia , Fibras Aferentes Viscerais/metabolismo
20.
Neurosci Lett ; 424(1): 10-5, 2007 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-17703884

RESUMO

The complement anaphylatoxin C5a is a potent mediator of the innate immune response to infection. Recent evidence also reveals that C5a contributes to central nervous system effects in addition to its well-known peripheral functions. However, it is not known if C5a has a role in the activation of the hypothalamic-pituitary-adrenal (HPA) axis; a critical cascade that exemplifies neuroimmune interactions between the periphery and the brain. In the present study we examined if systemic pre-treatment with a C5a receptor antagonist, PMX53, can affect lipopolysaccharide-induced (LPS; 1 mg/kg, i.p.) activation of the HPA axis in the rat. Using Fos protein as a marker of neuronal activation, we found that systemic administration of PMX53 reduced the LPS-induced activation of paraventricular corticotropin-releasing factor (PVN CRF) and central amygdala cells. However, PMX53 did not alter LPS-induced responses in the bed nucleus of the stria terminalis, nucleus tractus solitarius and ventrolateral medulla. Our findings demonstrate that C5a may have a role in the activation of the HPA axis in response to systemic LPS.


Assuntos
Tonsila do Cerebelo/imunologia , Complemento C5a/imunologia , Sistema Hipotálamo-Hipofisário/imunologia , Neuroimunomodulação/fisiologia , Núcleo Hipotalâmico Paraventricular/imunologia , Receptor da Anafilatoxina C5a/imunologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Hormônio Liberador da Corticotropina/imunologia , Hormônio Liberador da Corticotropina/metabolismo , Mediadores da Inflamação , Lipopolissacarídeos/farmacologia , Masculino , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Receptor da Anafilatoxina C5a/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA