Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 44(27)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38789262

RESUMEN

We previously showed that orexin neurons are activated by hypoxia and facilitate the peripheral chemoreflex (PCR)-mediated hypoxic ventilatory response (HVR), mostly by promoting the respiratory frequency response. Orexin neurons project to the nucleus of the solitary tract (nTS) and the paraventricular nucleus of the hypothalamus (PVN). The PVN contributes significantly to the PCR and contains nTS-projecting corticotropin-releasing hormone (CRH) neurons. We hypothesized that in male rats, orexin neurons contribute to the PCR by activating nTS-projecting CRH neurons. We used neuronal tract tracing and immunohistochemistry (IHC) to quantify the degree that hypoxia activates PVN-projecting orexin neurons. We coupled this with orexin receptor (OxR) blockade with suvorexant (Suvo, 20 mg/kg, i.p.) to assess the degree that orexin facilitates the hypoxia-induced activation of CRH neurons in the PVN, including those projecting to the nTS. In separate groups of rats, we measured the PCR following systemic orexin 1 receptor (Ox1R) blockade (SB-334867; 1 mg/kg) and specific Ox1R knockdown in PVN. OxR blockade with Suvo reduced the number of nTS and PVN neurons activated by hypoxia, including those CRH neurons projecting to nTS. Hypoxia increased the number of activated PVN-projecting orexin neurons but had no effect on the number of activated nTS-projecting orexin neurons. Global Ox1R blockade and partial Ox1R knockdown in the PVN significantly reduced the PCR. Ox1R knockdown also reduced the number of activated PVN neurons and the number of activated tyrosine hydroxylase neurons in the nTS. Our findings suggest orexin facilitates the PCR via nTS-projecting CRH neurons expressing Ox1R.


Asunto(s)
Hormona Liberadora de Corticotropina , Neuronas , Antagonistas de los Receptores de Orexina , Receptores de Orexina , Orexinas , Ratas Sprague-Dawley , Núcleo Solitario , Animales , Masculino , Hormona Liberadora de Corticotropina/metabolismo , Orexinas/metabolismo , Ratas , Neuronas/metabolismo , Neuronas/fisiología , Neuronas/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/fisiología , Núcleo Solitario/efectos de los fármacos , Antagonistas de los Receptores de Orexina/farmacología , Receptores de Orexina/metabolismo , Hipoxia/metabolismo , Triazoles/farmacología , Azepinas/farmacología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/fisiología
2.
J Neurosci ; 43(24): 4513-4524, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37160364

RESUMEN

Corticotropin-releasing hormone (CRH) is a neuropeptide regulating neuroendocrine and autonomic function. CRH mRNA and protein levels in the hypothalamic paraventricular nucleus (PVN) are increased in primary hypertension. However, the role of CRH in elevated sympathetic outflow in primary hypertension remains unclear. CRHR1 proteins were distributed in retrogradely labeled PVN presympathetic neurons with an increased level in the PVN tissue in adult spontaneously hypertensive rats (SHRs) compared with age-matched male Wistar-Kyoto (WKY) rats. CRH induced a more significant increase in the firing rate of PVN-rostral ventrolateral medulla (RVLM) neurons and sympathoexcitatory response in SHRs than in WKY rats, an effect that was blocked by preapplication of NMDA receptors (NMDARs) antagonist AP5 and PSD-95 inhibitor, Tat-N-dimer. Blocking CRHRs with astressin or CRHR1 with NBI35965 significantly decreased the firing rate of PVN-RVLM output neurons and reduced arterial blood pressure (ABP) and renal sympathetic nerve activity (RSNA) in SHRs but not in WKY, whereas blocking CRHR2 with antisauvagine-30 did not. Furthermore, Immunocytochemistry staining revealed that CRHR1 colocalized with NMDARs in PVN presympathetic neurons. Blocking CRHRs significantly decreased the NMDA currents in labeled PVN neurons. PSD-95-bound CRHR1 and PSD-95-bound GluN2A in the PVN were increased in SHRs. These data suggested that the upregulation of CRHR1 in the PVN is critically involved in the hyperactivity of PVN presympathetic neurons and elevated sympathetic outflow in primary hypertension.SIGNIFICANCE STATEMENT Our study found that corticotropin-releasing hormone receptor (CRHR)1 protein levels were increased in the paraventricular nucleus (PVN), and CRHR1 interacts with NMDA receptors (NMDARs) through postsynaptic density protein (PSD)-95 in the PVN neurons in primary hypertension. The increased CRHR1 and CRHR1-NMDAR-PSD-95 complex in the PVN contribute to the hyperactivity of the PVN presympathetic neurons and elevated sympathetic vasomotor tone in hypertension in SHRs. Thus, the antagonism of CRHR1 decreases sympathetic outflow and blood pressure in hypertension. These findings determine a novel role of CRHR1 in elevated sympathetic vasomotor tone in hypertension, which is useful for developing novel therapeutics targeting CRHR1 to treat elevated sympathetic outflow in primary hypertension. The CRHR1 receptor antagonists, which are used to treat health consequences resulting from chronic stress, are candidates to treat primary hypertension.


Asunto(s)
Hipertensión Esencial , Hipertensión , Receptores de N-Metil-D-Aspartato , Animales , Masculino , Ratas , Hormona Adrenocorticotrópica , Hormona Liberadora de Corticotropina/metabolismo , Hipertensión Esencial/metabolismo , Hipertensión/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Hormonas Liberadoras de Hormona Hipofisaria/metabolismo , Hormonas Liberadoras de Hormona Hipofisaria/farmacología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervioso Simpático/fisiología
3.
Arterioscler Thromb Vasc Biol ; 43(1): 92-108, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36412197

RESUMEN

BACKGROUND: Hematopoietic stem cell (HSC) therapy has shown promise for tissue regeneration after ischemia. Therefore, there is a need to understand mechanisms underlying endogenous HSCs activation in response to ischemic stress and coordination of angiogenesis and repair. SHP-1 plays important roles in HSC quiescence and differentiation by regulation of TGF-ß1 signaling. TGF-ß1 promotes angiogenesis by stimulating stem cells to secrete growth factors to initiate the formation of blood vessels and later aid in their maturation. We propose that SHP-1 responds to ischemia stress in HSC and progenitor cells (HSPC) via regulation of TGF-ß1. METHODS: A mouse hind limb ischemia model was used. Local blood perfusion in the limbs was determined using laser doppler perfusion imaging. The number of positive blood vessels per square millimeter, as well as blood vessel diameter (µm) and area (µm2), were calculated. Hematopoietic cells were analyzed using flow cytometry. The bone marrow transplantation assay was performed to measure HSC reconstitution. RESULTS: After femoral artery ligation, TGF-ß1 was initially decreased in the bone marrow by day 3 of ischemia, followed by an increase on day 7. This pattern was opposite to that in the peripheral blood, which is concordant with the response of HSC to ischemic stress. In contrast, SHP-1 deficiency in HSC is associated with irreversible activation of HSPCs in the bone marrow and increased circulating HSPCs in peripheral blood following limb ischemia. In addition, there was augmented auto-induction of TGF-ß1 and sustained inactivation of SHP-1-Smad2 signaling, which impacted TGF-ß1 expression in HSPCs in circulation. Importantly, restoration of normal T GF-ß1 oscillations helped in the recovery of limb repair and function. CONCLUSIONS: HSPC-SHP-1-mediated regulation of TGF-ß1 in both bone marrow and peripheral blood is required for a normal response to ischemic stress.


Asunto(s)
Células Madre Hematopoyéticas , Factor de Crecimiento Transformador beta1 , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Células Madre Hematopoyéticas/metabolismo , Isquemia , Extremidad Inferior
4.
BMC Endocr Disord ; 24(1): 45, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38622616

RESUMEN

BACKGROUND: Headache is a common occurrence after endoscopic endonasal surgery (EES) for pituitary adenomas and significantly impacts the quality of life of patients. This study aims to investigate the effectiveness of nasal irrigation in relieving postoperative headache after EES. METHODS: A retrospective analysis was conducted on a cohort of 101 patients (Cohort I) who underwent EES for pituitary adenomas to explore the risk factors associated with postoperative headache. Another cohort of 72 patients (Cohort II) who received adjuvant nasal irrigation following surgery was enrolled for further analysis. The Headache Impact Test (HIT-6) was used to score the severity of headache, and patients with a HIT score > 55 were classified as having headache. RESULTS: In Cohort I, 21.78% of patients experienced headache one month after EES, which decreased to 5.94% at the three-month follow-up. Multivariate analysis revealed that postoperative nasal sinusitis (OR = 3.88, 95%CI 1.16-13.03, p = 0.028) and Hardy's grade C-D (OR = 10.53, 95%CI 1.02-109.19, p = 0.049) independently predicted the presence of postoperative headache at one month. At the three-month follow-up, patients with sinusitis had higher HIT-6 scores compared to those without sinusitis (44.43 ± 9.78 vs. 39.72 ± 5.25, p = 0.017). In Cohort II, the incidence of sinusitis at three months was significantly lower than that in Cohort I (p = 0.028). Importantly, both the incidence of headache and HIT-6 scores in Cohort II were significantly lower than those in Cohort I at the one- and three-month follow-ups. CONCLUSIONS: Postoperative sinusitis is an independent risk factor for the development of headache following EES for pituitary adenomas. Prophylactic nasal irrigation helps relieve postoperative headache, possibly by preventing the occurrence of sinusitis.


Asunto(s)
Neoplasias Hipofisarias , Sinusitis , Humanos , Neoplasias Hipofisarias/cirugía , Estudios Retrospectivos , Calidad de Vida , Resultado del Tratamiento , Endoscopía/efectos adversos , Cefalea/etiología , Cefalea/prevención & control , Lavado Nasal (Proceso)
5.
Nano Lett ; 22(9): 3668-3677, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35439419

RESUMEN

The real-time monitoring of neurochemical release in vivo plays a critical role in understanding the biochemical process of the complex nervous system. Current technologies for such applications, including microdialysis and fast-scan cyclic voltammetry, suffer from limited spatiotemporal resolution or poor selectivity. Here, we report a soft implantable aptamer-graphene microtransistor probe for real-time monitoring of neurochemical release. As a demonstration, we show the monitoring of dopamine with nearly cellular-scale spatial resolution, high selectivity (dopamine sensor >19-fold over norepinephrine), and picomolar sensitivity, simultaneously. Systematic benchtop evaluations, ex vivo experiments, and in vivo studies in mice models highlight the key features and demonstrate the capability of capturing the dopamine release dynamics evoked by pharmacological stimulation, suggesting the potential applications in basic neuroscience studies and studying neurological disease-related processes. The developed system can be easily adapted for monitoring other neurochemicals and drugs by simply replacing the aptamers functionalized on the graphene microtransistors.


Asunto(s)
Dopamina , Grafito , Animales , Ratones , Norepinefrina , Oligonucleótidos
6.
J Neurosci ; 41(30): 6551-6563, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34193557

RESUMEN

The interplay between genetic and environmental factors is critically involved in hypertension development. The paraventricular nucleus (PVN) of the hypothalamus regulates sympathetic output during stress responses and chronic hypertension. In this study, we determined mechanisms of synaptic plasticity in the PVN in chronic stress-induced persistent hypertension in male borderline hypertensive rats (BHR), the first offspring of spontaneously hypertensive rats and normotensive Wistar-Kyoto rats. In Wistar-Kyoto rats, chronic unpredictable mild stress (CUMS) increased arterial blood pressure (ABP) and heart rate, which quickly returned to baseline after CUMS ended. In contrast, in BHR, CUMS caused persistent elevation in ABP, which lasted at least 2 weeks after CUMS ended. CUMS also increased the mRNA level of α2δ-1 and synaptic protein levels of GluN1, α2δ-1, and α2δ-1-GluN1 complexes in the PVN in BHR. Furthermore, CUMS significantly increased the frequency of miniature EPSCs and the amplitude of NMDAR currents in spinally projecting PVN neurons in BHR; these increases were normalized by blocking NMDARs with AP5, inhibiting α2δ-1 with gabapentin, or disrupting the α2δ-1-NMDAR interaction with α2δ-1Tat peptide. Microinjection of AP5 or α2δ-1Tat peptide into the PVN normalized elevated ABP and renal sympathetic nerve activity in stressed BHR. In addition, systemically administered gabapentin or memantine attenuated higher ABP induced by CUMS in BHR. Our findings indicate that chronic stress-induced persistent hypertension is mediated by augmented sympathetic outflow via α2δ-1-bound NMDARs in the PVN. This new information provides a cellular and molecular basis for how the genetic-environment interactions cause persistent hypertension.SIGNIFICANCE STATEMENT Chronic stress is a major risk factor for hypertension development, especially for individuals with a genetic predisposition to hypertension. Using a rat model of borderline hypertension, we showed that chronic stress induced long-lasting hypertension and sympathetic nerve hyperactivity, which were maintained by NMDAR activation in the hypothalamus. Chronic stress also increased the expression of α2δ-1, previously regarded as a Ca2+ channel subunit, promoting physical interaction with and synaptic trafficking of NMDARs in the hypothalamus. Inhibiting α2δ-1, blocking NMDARs, or disrupting α2δ-1-bound NMDARs reversed chronic stress-induced sympathetic outflow and persistent hypertension. Thus, α2δ-1-dependent NMDAR activity in the hypothalamus is an effector of genetic-environment interactions and may be targeted for treating stress-induced neurogenic hypertension.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Interacción Gen-Ambiente , Hipertensión/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Femenino , Predisposición Genética a la Enfermedad , Hipertensión/genética , Masculino , Plasticidad Neuronal/fisiología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Estrés Psicológico
7.
J Neurochem ; 161(6): 478-491, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35583089

RESUMEN

Glutamate N-methyl-d-aspartate (NMDA) receptors (NMDARs) and Kv7/M channels are importantly involved in regulating neuronal activity involved in various physiological and pathological functions. Corticotropin-releasing hormone (CRH)-expressing neurons in the central nucleus of the amygdala (CeA) critically mediate autonomic response during stress. However, the interaction between NMDA receptors and Kv7/M channels in the CRHCeA neurons remains unclear. In this study, we identified rat CRHCeA neurons through the expression of an AAV viral vector-mediated enhanced green fluorescent protein (eGFP) driven by the rat CRH promoter. M-currents carried by Kv7/M channels were recorded using the whole-cell patch-clamp approach in eGFP-tagged CRHCeA neurons in brain slices. Acute exposure to NMDA significantly reduced M-currents recorded from the CRHCeA neurons. NMDA-induced suppression of M-currents was eliminated by chelating intracellular Ca2+ , supplying phosphatidylinositol 4,5-bisphosphate (PIP2) intracellularly, or blocking phosphoinositide3-kinase (PI3K). In contrast, inhibiting protein kinase C (PKC) or calmodulin did not alter NMDA-induced suppression of M-currents. Sustained exposure of NMDA decreased Kv7.3 membrane protein levels and suppressed M-currents, while the Kv7.2 expression levels remained unaltered. Pre-treatment of brain slices with PKC inhibitors alleviated the decreases in Kv7.3 and reduction of M-currents in CRHCeA neurons induced by NMDA. PKC inhibitors did not alter Kv7.2 and Kv7.3 membrane protein levels and M-currents in CRHCeA neurons. These data suggest that transient activation of NMDARs suppresses M-currents through the Ca2+ -dependent PI3K-PIP2 signaling pathway. In contrast, sustained activation of NMDARs reduces Kv7.3 protein expression and suppresses M-currents through a PKC-dependent pathway.


Asunto(s)
Núcleo Amigdalino Central , Hormona Liberadora de Corticotropina , Animales , Hormona Liberadora de Corticotropina/metabolismo , N-Metilaspartato/farmacología , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología
8.
Neurobiol Dis ; 165: 105634, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35077822

RESUMEN

Neurocognitive consequences of blast-induced traumatic brain injury (bTBI) pose significant concerns for military service members and veterans with the majority of "invisible injury." However, the underlying mechanism of such mild bTBI by low-intensity blast (LIB) exposure for long-term cognitive and mental deficits remains elusive. Our previous studies have shown that mice exposed to LIB result in nanoscale ultrastructural abnormalities in the absence of gross or apparent cellular damage in the brain. Here we tested the hypothesis that glutamatergic hyperexcitability may contribute to long-term learning deficits. Using brain slice electrophysiological recordings, we found an increase in averaged frequencies with a burst pattern of miniature excitatory postsynaptic currents (mEPSCs) in hippocampal CA3 neurons in LIB-exposed mice at 1- and 7-days post injury, which was blocked by a specific NMDA receptor antagonist AP5. In addition, cognitive function assessed at 3-months post LIB exposure by automated home-cage monitoring showed deficits in dynamic patterns of discrimination learning and cognitive flexibility in LIB-exposed mice. Collected hippocampal tissue was further processed for quantitative global-proteomic analysis. Advanced data-independent acquisition for quantitative tandem mass spectrometry analysis identified altered expression of proteins involved in synaptic plasticity and serine protease inhibitors in LIB-exposed mice. Some were correlated with the ability of discrimination learning and cognitive flexibility. These findings show that acute glutamatergic hyperexcitability in the hippocampus induced by LIB may contribute to long-term cognitive dysfunction and protein alterations. Studies using this military-relevant mouse model of mild bTBI provide valuable insights into developing a potential therapeutic strategy to ameliorate hyperexcitability-modulated LIB injuries.


Asunto(s)
Traumatismos por Explosión , Proteómica , Animales , Traumatismos por Explosión/complicaciones , Traumatismos por Explosión/metabolismo , Hipocampo/metabolismo , Ratones , Plasticidad Neuronal , Inhibidores de Serina Proteinasa/metabolismo
9.
Int J Clin Oncol ; 27(9): 1386-1393, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35781641

RESUMEN

BACKGROUND: Cancer patients are associated with an elevated risk of suicide. This study aims to investigate the suicide rates and identify risk factors for suicide among patients with malignant intracranial tumors (MITs). METHODS: Patients diagnosed with MITs during the years of 1975-2015 were identified from the Surveillance, Epidemiology, and End Results (SEER) program. Suicide rates and standardized mortality ratios (SMR) were calculated. Cox regression analyses were used to identified risk factors for suicide among MIT patients. RESULTS: Among 115,668 patients with MITs collected from the SEER program, 99 committed suicide. The rate of suicide was 23.02 per 100,000 person-years, and SMR of suicide was 1.90. Diagnosis in recent era (years 2000-2015, SMR = 2.01), male gender (SMR = 1.78), older age (60-79 years, SMR = 3.54), white race (SMR = 1.86), married persons (SMR = 2.31), living in rural areas (SMR = 2.50), history of other malignancy (SMR = 3.81), diagnosis of glioblastoma (SMR = 4.05) and supratentorial location (SMR = 2.45) were associated with an increased incidence of suicide. In addition, the risk of suicide increased significantly within the first year after diagnosis (SMR = 13.04). Multivariate Cox regressions showed that older age, male sex, and supratentorial location were independent risk factors for suicide. CONCLUSIONS: The suicide mortality among patients with MITs steadily elevated in the past decades. Male sex, older age, and supratentorial location were significantly associated with risk of suicide, especially within the first year following diagnosis. Healthcare providers should early identify and effectively intervene with MIT patients at risk.


Asunto(s)
Neoplasias Encefálicas , Suicidio , Neoplasias Encefálicas/epidemiología , Humanos , Incidencia , Masculino , Factores de Riesgo , Programa de VERF
10.
BMC Endocr Disord ; 21(1): 225, 2021 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-34774043

RESUMEN

BACKGROUND: Prolactinoma is the major cause of hyperprolactinemia, and dopamine agonists (DAs) are generally the first-line treatment for them. Several studies have reviewed the recurrent rate of hyperprolactinemia after DAs withdrawal. However, few of them have concerned the recurrence risk of prolactinoma following the withdrawal of DAs. METHODS: Three medical databases, PubMed, EMBASE and Cochrane library, were retrieved up to February, 14, 2021 to identify studies related to recurrence of prolactinoma and withdrawal of DAs. Statistical analyses including meta-analysis, sensitivity analysis, meta-regression, funnel plot and Egger test were performed through software R. RESULTS: A total of 3225 studies were retrieved from the three data bases, and 13 studies consisted of 616 patients and 19 arms were finally included in this systematic analysis. There was no significant heterogeneity among the included studies, and fixed effect model was thus used. The pooled recurrence proportion of prolactinoma after withdrawal of DA was 2% with a 95% confidence interval (CI) of 1-3%. CONCLUSION: Our study showed a very low recurrent rate of prolactinomas after DAs withdrawal. Much more prospective studies with larger cases and longer follow-up period are encouraged to confirm our finding. TRIAL REGISTRATION: Registration number CRD42021245888 (PROSPERO).


Asunto(s)
Deprescripciones , Agonistas de Dopamina/uso terapéutico , Recurrencia Local de Neoplasia/epidemiología , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactinoma/tratamiento farmacológico , Bromocriptina/uso terapéutico , Cabergolina/uso terapéutico , Humanos
11.
J Neurosci ; 38(28): 6388-6398, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29921713

RESUMEN

Both the sympathetic nervous system and the renin-angiotensin system are critically involved in hypertension development. Although angiotensin II (Ang II) stimulates hypothalamic paraventricular nucleus (PVN) neurons to increase sympathetic vasomotor tone, the molecular mechanism mediating this action remains unclear. The glutamate NMDAR in the PVN controls sympathetic outflow in hypertension. In this study, we determined the interaction between α2δ-1 (encoded by Cacna2d1), commonly known as a Ca2+ channel subunit, and NMDARs in the hypothalamus and its role in Ang II-induced synaptic NMDAR activity in PVN presympathetic neurons. Coimmunoprecipitation assays showed that α2δ-1 interacted with the NMDAR in the hypothalamus of male rats and humans (both sexes). Ang II increased the prevalence of synaptic α2δ-1-NMDAR complexes in the hypothalamus. Also, Ang II increased presynaptic and postsynaptic NMDAR activity via AT1 receptors, and such effects were abolished either by treatment with pregabalin, an inhibitory α2δ-1 ligand, or by interrupting the α2δ-1-NMDAR interaction with an α2δ-1 C terminus-interfering peptide. In Cacna2d1 knock-out mice (both sexes), Ang II failed to affect the presynaptic and postsynaptic NMDAR activity of PVN neurons. In addition, the α2δ-1 C terminus-interfering peptide blocked the sympathoexcitatory response to microinjection of Ang II into the PVN. Our findings indicate that Ang II augments sympathetic vasomotor tone and excitatory glutamatergic input to PVN presympathetic neurons by stimulating α2δ-1-bound NMDARs at synapses. This information extends our understanding of the molecular basis for the interaction between the sympathetic nervous and renin-angiotensin systems and suggests new strategies for treating neurogenic hypertension.SIGNIFICANCE STATEMENT Although both the sympathetic nervous system and renin-angiotensin system are closely involved in hypertension development, the molecular mechanisms mediating this involvement remain unclear. We showed that α2δ-1, previously known as a calcium channel subunit, interacts with NMDARs in the hypothalamus of rodents and humans. Angiotensin II (Ang II) increases the synaptic expression level of α2δ-1-NMDAR complexes. Furthermore, inhibiting α2δ-1, interrupting the α2δ-1-NMDAR interaction, or deleting α2δ-1 abolishes the potentiating effects of Ang II on presynaptic and postsynaptic NMDAR activity in the hypothalamus. In addition, the sympathoexcitatory response to Ang II depends on α2δ-1-bound NMDARs. Thus, α2δ-1-NMDAR complexes in the hypothalamus serve as an important molecular substrate for the interaction between the sympathetic nervous system and the renin-angiotensin system. This evidence suggests that α2δ-1 may be a useful target for the treatment neurogenic hypertension.


Asunto(s)
Angiotensina II/metabolismo , Canales de Calcio/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Renina-Angiotensina/fisiología , Sistema Nervioso Simpático/fisiología , Angiotensina II/farmacología , Animales , Femenino , Humanos , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
12.
J Biol Chem ; 293(50): 19354-19364, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30355732

RESUMEN

The striatum receives extensive cortical input and plays a prominent role in motor learning and habit formation. Glutamate N-methyl-d-aspartate (NMDA) receptor (NMDAR)-mediated long-term potentiation (LTP) is a major synaptic plasticity involved in learning and memory. However, the molecular mechanism underlying NMDAR plasticity in corticostriatal LTP is unclear. Here, we show that theta-burst stimulation (TBS) consistently induced corticostriatal LTP and increased the coincident presynaptic and postsynaptic NMDAR activity of medium spiny neurons. We also found that α2δ-1 (previously known as a subunit of voltage-gated calcium channels; encoded by the Cacna2d1 gene) physically interacted with NMDARs in the striatum of mice and humans, indicating that this cross-talk is conserved across species. Strikingly, inhibiting α2δ-1 trafficking with gabapentin or disrupting the α2δ-1-NMDAR interaction with an α2δ-1 C terminus-interfering peptide abolished TBS-induced LTP. In Cacna2d1-knockout mice, TBS failed to induce corticostriatal LTP and the associated increases in presynaptic and postsynaptic NMDAR activities. Moreover, systemic gabapentin treatment, microinjection of α2δ-1 C terminus-interfering peptide into the dorsomedial striatum, or Cacna2d1 ablation impaired the alternation T-maze task and rotarod performance in mice. Our findings indicate that the interaction between α2δ-1 and NMDARs is of high physiological relevance and that a TBS-induced switch from α2δ-1-free to α2δ-1-bound NMDARs is critically involved in corticostriatal LTP and LTP-associated learning and memory. Gabapentinoids at high doses may adversely affect cognitive function by targeting α2δ-1-NMDAR complexes.


Asunto(s)
Canales de Calcio/metabolismo , Potenciación a Largo Plazo , Memoria/fisiología , Neostriado/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Femenino , Masculino , Memoria a Corto Plazo , Ratones , Actividad Motora/fisiología , Neostriado/citología , Neostriado/metabolismo , Unión Proteica , Aprendizaje Espacial , Sinapsis/metabolismo
13.
J Neurooncol ; 145(1): 125-134, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31493161

RESUMEN

PURPOSE: We aim to investigate the impacts of extent of resection and adjuvant radiotherapy on survival of high-grade meningiomas (WHO grade II-III) according to modern diagnosis and management. METHODS: Patients with high-grade meningiomas were identified in the Surveillance Epidemiology and End Results (SEER) database between 2000 and 2015 and used for survival analysis. Propensity score matching (PSM) was conducted to reduce selection bias. Another 92 patients from Sun Yat-sen University Cancer Center (SYSUCC) were used for validation. RESULTS: 530 patients were enrolled from SEER. Patients with gross total resection (GTR) had no significantly different overall survival (OS) compared with those with subtotal resection (STR), even after performing PSM between these two groups. Multivariable analysis found that age ≥ 65 years (HR 2.22, P < 0.001), tumor diameter > 6 cm (HR 1.59, P = 0.004) and grade III tumor (HR 4.31, P < 0.001) were associated with worse OS. Stratification analysis showed that adjuvant radiotherapy conferred significantly improved OS for grade III meningiomas, but not for grade II meningiomas, regardless of resection extent. In SYSUCC cohort, resection extent was also not significantly associated with OS. However, patients with GTR (Simpson grade I-III) had distinctly increased progression-free survival (PFS) than those with STR (P < 0.001). Additionally, for grade II meningiomas after GTR, radiotherapy was unable to improve OS and PFS. CONCLUSION: On modern management of high-grade meningiomas, GTR does not improve OS, but seems to be associated with increased PFS. Radiotherapy is reasonable as a supplement for treating grade III meningiomas, whereas its effect for grade II meningiomas remains uncertain and needs further validation by prospective study.


Asunto(s)
Neoplasias Meníngeas/mortalidad , Meningioma/mortalidad , Procedimientos Neuroquirúrgicos/mortalidad , Radioterapia Adyuvante/mortalidad , Adolescente , Adulto , Anciano , Estudios de Cohortes , Terapia Combinada , Manejo de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/terapia , Meningioma/patología , Meningioma/terapia , Persona de Mediana Edad , Clasificación del Tumor , Tasa de Supervivencia , Adulto Joven
14.
J Neurosci ; 37(44): 10690-10699, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28972129

RESUMEN

NMDAR activity in the hypothalamic paraventricular nucleus (PVN) is increased and critically involved in heightened sympathetic vasomotor tone in hypertension. Calcium/calmodulin-dependent protein kinase II (CaMKII) binds to and modulates NMDAR activity. In this study, we determined the role of CaMKII in regulating NMDAR activity of PVN presympathetic neurons in male spontaneously hypertensive rats (SHRs). NMDAR-mediated EPSCs and puff NMDA-elicited currents were recorded in spinally projecting PVN neurons in SHRs and male Wistar-Kyoto (WKY) rats. The basal amplitude of evoked NMDAR-EPSCs and puff NMDA currents in retrogradely labeled PVN neurons were significantly higher in SHRs than in WKY rats. The CaMKII inhibitor autocamtide-2-related inhibitory peptide (AIP) normalized the increased amplitude of NMDAR-EPSCs and puff NMDA currents in labeled PVN neurons in SHRs but had no effect in WKY rats. Treatment with AIP also normalized the higher frequency of NMDAR-mediated miniature EPSCs of PVN neurons in SHRs. CaMKII-mediated phosphorylation level of GluN2B serine 1303 (S1303) in the PVN, but not in the hippocampus and frontal cortex, was significantly higher in SHRs than in WKY rats. Lowering blood pressure with celiac ganglionectomy in SHRs did not alter the increased level of phosphorylated GluN2B S1303 in the PVN. In addition, microinjection of AIP into the PVN significantly reduced arterial blood pressure and lumbar sympathetic nerve discharges in SHRs. Our findings suggest that CaMKII activity is increased in the PVN and contributes to potentiated presynaptic and postsynaptic NMDAR activity to elevate sympathetic vasomotor tone in hypertension.SIGNIFICANCE STATEMENT Heightened sympathetic vasomotor tone is a major contributor to the development of hypertension. Although glutamate NMDA receptor (NMDAR)-mediated excitatory drive in the hypothalamus plays a critical role in increased sympathetic output in hypertension, the molecular mechanism involved in potentiated NMDAR activity of hypothalamic presympathetic neurons remains unclear. Here we show that the activity of calcium/calmodulin-dependent protein kinase II (CaMKII) is increased and plays a key role in the potentiated presynaptic and postsynaptic NMDAR activity of hypothalamic presympathetic neurons in hypertension. Also, the inhibition of CaMKII in the hypothalamus reduces elevated blood pressure and sympathetic nerve discharges in hypertension. This new knowledge extends our understanding of the mechanism of synaptic plasticity in the hypothalamus and suggests new strategies to treat neurogenic hypertension.


Asunto(s)
Fibras Adrenérgicas/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Hipertensión/fisiopatología , Hipotálamo/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de N-Metil-D-Aspartato
15.
J Physiol ; 596(17): 4269-4283, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29971791

RESUMEN

KEY POINTS: α2δ-1 is upregulated, promoting the interaction with NMDA receptors (NMDARs), in the hypothalamus in a rat model of hypertension. The prevalence of α2δ-1-bound NMDARs at synaptic sites in the hypothalamus is increased in hypertensive animals. α2δ-1 is essential for the increased presynaptic and postsynaptic NMDAR activity of hypothalamic neurons in hypertension. α2δ-1-bound NMDARs in the hypothalamus are critically involved in augmented sympathetic outflow in hypertensive animals. ABSTRACT: Increased glutamate NMDA receptor (NMDAR) activity in the paraventricular nucleus (PVN) of the hypothalamus leads to augmented sympathetic outflow in hypertension. However, the molecular mechanisms underlying this effect remain unclear. α2δ-1, previously considered to be a voltage-activated calcium channel subunit, is a newly discovered powerful regulator of NMDARs. In the present study, we determined the role of α2δ-1 in regulating synaptic NMDAR activity of rostral ventrolateral medulla (RVLM)-projecting PVN neurons in spontaneously hypertensive rats (SHRs). We show that the protein levels of α2δ-1 and NMDARs in synaptosomes and the α2δ-1-NMDAR complexes in the hypothalamus were substantially higher in SHRs than in normotensive control rats. The basal amplitude of evoked NMDAR currents and NMDAR-mediated synaptic glutamate release in RVLM-projecting PVN neurons were significantly increased in SHRs. Strikingly, inhibiting α2δ-1 activity with gabapentin or disrupting the α2δ-1-NMDAR association with an α2δ-1 C-terminus peptide completely normalized the amplitude of evoked NMDAR currents and NMDAR-mediated synaptic glutamate release in RVLM-projecting PVN neurons in SHRs. In addition, microinjection of the α2δ-1 C-terminus peptide into the PVN substantially reduced arterial blood pressure and renal sympathetic nerve discharges in SHRs. Our findings indicate that α2δ-1-bound NMDARs in the PVN are required for the potentiated presynaptic and postsynaptic NMDAR activity of PVN presympathetic neurons and for the elevated sympathetic outflow in hypertension. α2δ-1-bound NMDARs may be an opportune target for treating neurogenic hypertension.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Potenciales Postsinápticos Excitadores , Hipertensión/fisiopatología , Hipotálamo/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiopatología , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervioso Simpático/fisiopatología , Animales , Ácido Glutámico/metabolismo , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
16.
Am J Physiol Heart Circ Physiol ; 315(5): H1200-H1214, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30095973

RESUMEN

The hypothalamic paraventricular nucleus (PVN) is a unique and important brain region involved in the control of cardiovascular, neuroendocrine, and other physiological functions pertinent to homeostasis. The PVN is a major source of excitatory drive to the spinal sympathetic outflow via both direct and indirect projections. In this review, we discuss the role of the PVN in the regulation of sympathetic output in normal physiological conditions and in hypertension. In normal healthy animals, the PVN presympathetic neurons do not appear to have a major role in sustaining resting sympathetic vasomotor activity or in regulating sympathetic responses to short-term homeostatic challenges such as acute hypotension or hypoxia. Their role is, however, much more significant during longer-term challenges, such as sustained water deprivation, chronic intermittent hypoxia, and pregnancy. The PVN also appears to have a major role in generating the increased sympathetic vasomotor activity that is characteristic of multiple forms of hypertension. Recent studies in the spontaneously hypertensive rat model have shown that impaired inhibitory and enhanced excitatory synaptic inputs to PVN presympathetic neurons are the basis for the heightened sympathetic outflow in hypertension. We discuss the molecular mechanisms underlying the presynaptic and postsynaptic alterations in GABAergic and glutamatergic inputs to PVN presympathetic neurons in hypertension. In addition, we discuss the ability of exercise training to correct sympathetic hyperactivity by restoring blood-brain barrier integrity, reducing angiotensin II availability, and decreasing oxidative stress and inflammation in the PVN.


Asunto(s)
Presión Sanguínea , Sistema Cardiovascular/inervación , Hipertensión/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiopatología , Transmisión Sináptica , Sistema Vasomotor/fisiopatología , Angiotensina II/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores , Terapia por Ejercicio , Neuronas GABAérgicas/metabolismo , Ácido Glutámico/metabolismo , Humanos , Hipertensión/metabolismo , Hipertensión/terapia , Mediadores de Inflamación/metabolismo , Inhibición Neural , Estrés Oxidativo , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Vasomotor/metabolismo
17.
J Neurochem ; 142(4): 512-520, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28547758

RESUMEN

Ghrelin increases food intake and body weight by stimulating orexigenic agouti-related protein (AgRP)/neuropeptide Y (NPY) neurons and inhibiting anorexic pro-opiomelanocortin (POMC) neurons in the hypothalamus. Growth hormone secretagogue receptor (Ghsr) mediates the effect of ghrelin on feeding behavior and energy homeostasis. However, the role of Ghsr in the ghrelin effect on these two populations of neurons is unclear. We hypothesized that Ghsr mediates the effect of ghrelin on AgRP and POMC neurons. In this study, we determined whether Ghsr similarly mediates the effects of ghrelin on AgRP/NPY and POMC neurons using cell type-specific Ghsr-knockout mice. Perforated whole-cell recordings were performed on green fluorescent protein-tagged AgRP/NPY and POMC neurons in the arcuate nucleus in hypothalamic slices. In Ghsr+/+ mice, ghrelin (100 nM) significantly increased the firing activity of AgRP/NPY neurons but inhibited the firing activity of POMC neurons. In Ghsr-/- mice, the excitatory effect of ghrelin on AgRP/NPY neurons was abolished. Ablation of Ghsr also eliminated ghrelin-induced increases in the frequency of GABAergic inhibitory postsynaptic currents of POMC neurons. Strikingly, ablation of Ghsr converted the ghrelin effect on POMC neurons from inhibition to excitation. Des-acylated ghrelin had no such effect on POMC neurons in Ghsr-/- mice. In both Ghsr+/+ and Ghsr-/- mice, blocking GABAA receptors with gabazine increased the basal firing activity of POMC neurons, and ghrelin further increased the firing activity of POMC neurons in the presence of gabazine. Our findings provide unequivocal evidence that Ghsr is essential for ghrelin-induced excitation of AgRP/NPY neurons. However, ghrelin excites POMC neurons through an unidentified mechanism that is distinct from conventional Ghsr.


Asunto(s)
Ghrelina/farmacología , Hipotálamo/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Receptores de Ghrelina/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ratones Transgénicos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Proopiomelanocortina/metabolismo
18.
Am J Physiol Regul Integr Comp Physiol ; 312(6): R982-R995, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28404583

RESUMEN

Hypoxia results in decreased arterial Po2, arterial chemoreflex activation, and compensatory increases in breathing, sympathetic outflow, and neuroendocrine secretions, including increased secretion of AVP, corticotropin-releasing hormone (CRH), adrenocorticotropin hormone (ACTH), and corticosterone. In addition to a brain stem pathway, including the nucleus tractus solitarius (nTS) and the rostral ventrolateral medulla (RVLM), medullary pathways to the paraventricular nucleus of the hypothalamus (PVN) contribute to chemoreflex responses. Experiments evaluated activation of specific cell phenotypes within the PVN following an acute hypoxic stimulus (AH; 2 h, 10% O2) in conscious rats. Retrograde tracers (from spinal cord and RVLM) labeled presympathetic (PreS) neurons, and immunohistochemistry identified AVP- and CRH-immunoreactive (IR) cells. c-Fos-IR was an index of neuronal activation. Hypoxia activated AVP-IR (~6%) and CRH-IR (~15%) cells, but not PreS cells in the PVN, suggesting that sympathoexcitation during moderate AH is mediated mainly by a pathway that does not include PreS neurons in the PVN. Approximately 14 to 17% of all PVN cell phenotypes examined expressed neuronal nitric oxide synthase (nNOS-IR). AH activated only nNOS-negative AVP-IR neurons. In contrast ~23% of activated CRH-IR neurons in the PVN contained nNOS. In the median eminence, CRH-IR terminals were closely opposed to tanycyte processes and end-feet (vimentin-IR) in the external zone, where vascular NO participates in tanycyte retraction to facilitate neuropeptide secretion into the pituitary portal circulation. Results are consistent with an inhibitory role of NO on AVP and PreS neurons in the PVN and an excitatory role of NO on CRH secretion in the PVN and median eminence.


Asunto(s)
Fibras Adrenérgicas/metabolismo , Arginina Vasopresina/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Hipoxia/metabolismo , Células Neuroendocrinas/metabolismo , Neuronas Nitrérgicas/metabolismo , Óxido Nítrico/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Células Ependimogliales/metabolismo , Hipoxia/fisiopatología , Masculino , Inhibición Neural , Óxido Nítrico Sintasa de Tipo I/metabolismo , Núcleo Hipotalámico Paraventricular/fisiopatología , Fenotipo , Ratas Sprague-Dawley , Vimentina/metabolismo
19.
Neuroendocrinology ; 104(2): 194-208, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27077366

RESUMEN

INTRODUCTION: Prolonged and repeated stresses cause hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis. The corticotrophin-releasing hormone (CRH)-expressing neurons in the hypothalamic paraventricular nucleus (PVN) are an essential component of the HPA axis. MATERIALS AND METHODS: Chronic unpredictable mild stress (CUMS) was induced in Sprague-Dawley rats. GABA reversal potentials (EGABA) were determined by using gramicidin-perforated recordings in identified PVN-CRH neurons through expressing enhanced green fluorescent protein driven by the CRH promoter. Plasma corticosterone (CORT) levels were measured in rats implanted with a cannula targeting the lateral ventricles and PVN. RESULTS: Blocking the GABAA receptor in the PVN with gabazine significantly increased plasma CORT levels in unstressed rats but did not change CORT levels in CUMS rats. CUMS caused a depolarizing shift in EGABA in PVN-CRH neurons compared with EGABA in PVN-CRH neurons in unstressed rats. Furthermore, CUMS induced a long-lasting increase in expression levels of the cation chloride cotransporter Na+-K+-Cl--Cl- (NKCC1) in the PVN but a transient decrease in expression levels of K+-Cl--Cl- in the PVN, which returned to the basal level 5 days after CUMS treatment. The NKCC1 inhibitor bumetanide decreased the basal firing activity of PVN-CRH neurons and normalized EGABA and the gabazine-induced excitatory effect on PVN-CRH neurons in CUMS rats. In addition, central administration of bumetanide decreased basal circulating CORT levels in CUMS rats. CONCLUSIONS: These data suggest that chronic stress impairs GABAergic inhibition, resulting in HPA axis hyperactivity through upregulation of NKCC1.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Inhibición Neural , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiopatología , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Estrés Psicológico/fisiopatología , Ácido gamma-Aminobutírico/fisiología , Animales , Bumetanida/administración & dosificación , Corticosterona/sangre , Agonistas de Receptores de GABA-A/administración & dosificación , Antagonistas de Receptores de GABA-A/administración & dosificación , Masculino , Potenciales de la Membrana/efectos de los fármacos , Muscimol/administración & dosificación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Piridazinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/fisiología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/administración & dosificación , Estrés Psicológico/metabolismo , Regulación hacia Arriba
20.
Curr Hypertens Rep ; 19(10): 78, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28929331

RESUMEN

Elevated sympathetic vasomotor tone emanating from the brain is a major mechanism involved in the development of hypertension. Increased glutamatergic excitatory input to presympathetic neurons in the paraventricular nucleus (PVN) of the hypothalamus leads to increased sympathetic outflow in various animal models of hypertension. Recent studies have revealed molecular and cellular mechanisms underlying enhanced glutamatergic synaptic input to PVN presympathetic neurons in hypertension. In this review article, we summarize recent findings on changes in inotropic and metabotropic glutamate receptors, at both presynaptic and postsynaptic sites, responsible for increased glutamatergic input to PVN presympathetic neurons in hypertension. Particular emphasis is placed on the role of protein kinases and phosphatases in the potentiated activity of synaptic NMDA receptors in the PVN in hypertension. New findings about glutamatergic synaptic plasticity in the PVN not only improve the understanding of molecular mechanisms involved in heightened activity of the sympathetic nervous system but also suggest new therapeutic targets for treating drug-resistant, neurogenic hypertension.


Asunto(s)
Hipertensión/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiopatología , Receptores de Glutamato/metabolismo , Sistema Nervioso Simpático/fisiopatología , Animales , Ácido Glutámico/metabolismo , Humanos , Hipotálamo/fisiopatología , Plasticidad Neuronal , Neuronas/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA