scholarly journals Pro‐inflammatory cytokines increase neuronal activity in the hypothalamic paraventricular nucleus and contribute to the pathogenesis of streptozotocin‐induced diabetes

2009 ◽  
Vol 23 (S1) ◽  
Author(s):  
Yu‐Ming Kang ◽  
Da‐Nian Qin ◽  
Carrie Elks ◽  
Joseph Francis
2020 ◽  
Vol 17 (1) ◽  
Author(s):  
Ferdinand Althammer ◽  
Hildebrando Candido Ferreira-Neto ◽  
Myurajan Rubaharan ◽  
Ranjan K. Roy ◽  
Atit A. Patel ◽  
...  

Abstract Background Cardiovascular diseases, including heart failure, are the most common cause of death globally. Recent studies support a high degree of comorbidity between heart failure and cognitive and mood disorders resulting in memory loss, depression, and anxiety. While neuroinflammation in the hypothalamic paraventricular nucleus contributes to autonomic and cardiovascular dysregulation in heart failure, mechanisms underlying cognitive and mood disorders in this disease remain elusive. The goal of this study was to quantitatively assess markers of neuroinflammation (glial morphology, cytokines, and A1 astrocyte markers) in the central amygdala, a critical forebrain region involved in emotion and cognition, and to determine its time course and correlation to disease severity during the progression of heart failure. Methods We developed and implemented a comprehensive microglial/astrocyte profiler for precise three-dimensional morphometric analysis of individual microglia and astrocytes in specific brain nuclei at different time points during the progression of heart failure. To this end, we used a well-established ischemic heart failure rat model. Morphometric studies were complemented with quantification of various pro-inflammatory cytokines and A1/A2 astrocyte markers via qPCR. Results We report structural remodeling of central amygdala microglia and astrocytes during heart failure that affected cell volume, surface area, filament length, and glial branches, resulting overall in somatic swelling and deramification, indicative of a change in glial state. These changes occurred in a time-dependent manner, correlated with the severity of heart failure, and were delayed compared to changes in the hypothalamic paraventricular nucleus. Morphometric changes correlated with elevated mRNA levels of pro-inflammatory cytokines and markers of reactive A1-type astrocytes in the paraventricular nucleus and central amygdala during heart failure. Conclusion We provide evidence that in addition to the previously described hypothalamic neuroinflammation implicated in sympathohumoral activation during heart failure, microglia, and astrocytes within the central amygdala also undergo structural remodeling indicative of glial shifts towards pro-inflammatory phenotypes. Thus, our studies suggest that neuroinflammation in the amygdala stands as a novel pathophysiological mechanism and potential therapeutic target that could be associated with emotional and cognitive deficits commonly observed at later stages during the course of heart failure.


Author(s):  
Ferdinand Althammer ◽  
Hildebrando Candido Ferreira-Neto ◽  
Myurajan Rubaharan ◽  
Ranjan Kumer Roy ◽  
javier E Stern

Abstract Background Cardiovascular diseases, including heart failure are the most common cause of death globally. Recent studies support a high degree of comorbidity between heart failure and cognitive and mood disorders resulting in memory loss, depression and anxiety. While neuroinflammation in the hypothalamic paraventricular nucleus contributes to autonomic and cardiovascular dysregulation in heart failure, mechanisms underlying cognitive and mood disorders in this disease remain elusive. The goal of this study was to quantitatively asses markers of neuroinflammation (glial morphology, cytokines and A1 astrocyte markers) in the central amygdala, a critical forebrain region involved in emotion and cognition, and to determine its time course and correlation to disease severity during the progression of heart failure.Methods We developed and implemented a comprehensive microglial/astrocyte profiler for precise three-dimensional morphometric analysis of individual microglia and astrocytes in specific brain nuclei at different time points during the progression of heart failure. To this end, we used a well-established ischemic heart failure rat model. Morphometric studies were complemented with quantification of various pro-inflammatory cytokines and A1/A2 astrocyte markers via qPCR. Results We report structural remodeling of central amygdala microglia and astrocytes during heart failure that affected cell volume, surface area, filament length and microglial branches, resulting overall in somatic swelling and deramification, indicative of a change in microglial state. These changes occurred in a time-dependent manner, correlated with the severity of heart failure, and were delayed compared to changes in the hypothalamic paraventricular nucleus. Morphometric changes correlated with elevated mRNA levels of pro-inflammatory cytokines and markers of reactive A1-type astrocytes in the paraventricular nucleus and central amygdala during heart failure. Conclusion We provide evidence that in addition to the previously described hypothalamic neuroinflammation implicated in sympathohumoral activation during heart failure, microglia and astrocytes within the central amygdala also undergo structural remodeling indicative of glial shifts towards activated and pro-inflammatory phenotypes, respectively. Thus, our studies suggest that neuroinflammation in the amygdala stands as a novel pathophysiological mechanism and potential therapeutic target for that could be associated with emotional and cognitive deficits commonly observed at later stages during the course of heart failure.


2013 ◽  
Vol 305 (12) ◽  
pp. H1772-H1780 ◽  
Author(s):  
Amanda L. Sharpe ◽  
Alfredo S. Calderon ◽  
Mary Ann Andrade ◽  
J. Thomas Cunningham ◽  
Steven W. Mifflin ◽  
...  

Like humans with sleep apnea, rats exposed to chronic intermittent hypoxia (CIH) experience arterial hypoxemias and develop hypertension characterized by exaggerated sympathetic nerve activity (SNA). To gain insights into the poorly understood mechanisms that initiate sleep apnea/CIH-associated hypertension, experiments were performed in rats exposed to CIH for only 7 days. Compared with sham-treated normoxic control rats, CIH-exposed rats ( n = 8 rats/group) had significantly increased hematocrit ( P < 0.001) and mean arterial pressure (MAP; P < 0.05). Blockade of ganglionic transmission caused a significantly ( P < 0.05) greater reduction of MAP in rats exposed to CIH than control rats ( n = 8 rats/group), indicating a greater contribution of SNA in the support of MAP even at this early stage of CIH hypertension. Chemical inhibition of neuronal discharge in the hypothalamic paraventricular nucleus (PVN) (100 pmol muscimol) had no effect on renal SNA but reduced lumbar SNA ( P < 0.005) and MAP ( P < 0.05) more in CIH-exposed rats ( n = 8) than control rats ( n = 7), indicating that CIH increased the contribution of PVN neuronal activity in the support of lumbar SNA and MAP. Because CIH activates brain regions controlling body fluid homeostasis, the effects of internal carotid artery injection of hypertonic saline were tested and determined to increase lumbar SNA more ( P < 0.05) in CIH-exposed rats than in control rats ( n = 9 rats/group). We conclude that neurogenic mechanisms are activated early in the development of CIH hypertension such that elevated MAP relies on increased sympathetic tonus and ongoing PVN neuronal activity. The increased sensitivity of Na+/osmosensitive circuitry in CIH-exposed rats suggests that early neuroadaptive responses among body fluid regulatory neurons could contribute to the initiation of CIH hypertension.


2021 ◽  
Vol 15 ◽  
Author(s):  
Xiao-Jing Yu ◽  
Tong Xiao ◽  
Xiao-Jing Liu ◽  
Ying Li ◽  
Jie Qi ◽  
...  

The incidence rate and mortality of hypertension increase every year. Hypothalamic paraventricular nucleus (PVN) plays a critical role on the pathophysiology of hypertension. It has been demonstrated that the imbalance of neurotransmitters including norepinephrine (NE), glutamate (Glu) and γ-aminobutyric acid (GABA) are closely related to sympathetic overactivity and pathogenesis of hypertension. N-methyl-D-aspartate receptor (NMDAR), consisting of GluN1 and GluN2 subunits, is considered to be a glutamate-gated ion channel, which binds to Glu, and activates neuronal activity. Studies have found that the synthesis of respiratory chain enzyme complex was affected and mitochondrial function was impaired in spontaneously hypertensive rats (SHR), further indicating that mitochondria is associated with hypertension. Nuclear respiratory factor 1 (Nrf1) is a transcription factor that modulates mitochondrial respiratory chain and is related to GluN1, GluN2A, and GluN2B promoters. However, the brain mechanisms underlying PVN Nrf1 modulating sympathoexcitation and blood pressure during the development of hypertension remains unclear. In this study, an adeno-associated virus (AAV) vector carrying the shRNA targeting rat Nrf1 gene (shNrf1) was injected into bilateral PVN of male rats underwent two kidneys and one clip to explore the role of Nrf1 in mediating the development of hypertension and sympathoexcitation. Administration of shNrf1 knocked down the expression of Nrf1 and reduced the expression of excitatory neurotransmitters, increased the expression of inhibitory neurotransmitters, and reduced the production of reactive oxygen species (ROS), and attenuated sympathoexcitation and hypertension. The results indicate that knocking down Nrf1 suppresses sympathoexcitation in hypertension by reducing PVN transcription of NMDAR subunits (GluN1, GluN2A, and GluN2B), rebalancing PVN excitatory and inhibitory neurotransmitters, inhibiting PVN neuronal activity and oxidative stress, and attenuating sympathetic activity.


Hypertension ◽  
2012 ◽  
Vol 60 (suppl_1) ◽  
Author(s):  
Gustavo S Masson ◽  
Tassia S Costa ◽  
Lisete C Michelini

Aim: To analyze the sequence of adaptive responses induced by training (T) on both hemodynamic profile and cytokines gene expression within the paraventricular nucleus (PVN). Methods: SHR submitted to T (treadmill, 55% maximal capacity, 1h/day, 5 d/week) or kept sedentary (S), were cannulated for measurement of pressure (BP), heart rate (HR) and baroreflex sensitivity (BrS) at rest on weeks 0, 1, 2, 4 and 8 of protocols. HR and BP variability (power spectral analysis) were also evaluated. After euthanasia, brains were removed for isolation of PVN. Total mRNA was extracted for quantification of TNFα and IL-6 expression by real time PCR (HPRT as the endogenous gene). WKY were used as time controls. One-way ANOVA, with Fisher post-hoc test and Pearson correlation were employed. Results: At the beginning of the protocols, SHR exhibited high mean BP and HR, decreased BrS, increased BP variability and decreased HR variability (170±3 mmHg, 356±7 b/min; 2.3±0.2 b/min/mmHg, 14±2 mmHg 2 and 14±2 ms 2 ) when compared to WKY (119±2 mmHg, 307±6 b/min; 3.6±0.3 b/min/mmHg, 6±1 mmHg 2 and 22±2 ms 2 , respectively). Pro-inflammatory cytokines expression in PVN are also elevated in SHR vs WKY (TNF-α= 1.9±0.3 vs 1.2±0.1, IL-6= 4.9±0.8 vs 1.1±0.3 UA). In the SHR, T caused significant increases on BrS (4.0±0.2 b/min/mmHg, T 2 -T 8 ), HR variability (21±1 ms 2 , with a 60% increase of HF component from T 4 -T 8 ) and prevented the increase on BP variability in the SHR (13±2 vs 20±3 mmHg 2 , T 8 vs S 8 , with 29% reduction of LF component at T 8 ). T caused a prompt normalization of TNFα and IL-6 mRNA expression within the PVN (1.3±0.2 and 0.9±2 UA at T 2 ), accompanied by HR reduction (338±4 b/min, T 4 -T 8 ) and mean BP fall (159±6 mmHg, T 8 ), with significant correlations between TNFα x HR variability (Y=-3.6x+24.3, r= -0.29; P=0.04), TNFα x BrS (Y=-0.7x+4.3, r=-0.35; P=0.01), TNFα x BP (Y=9x+140, r= 0.26; P=0.04) and TNFα x HR (Y=18x+309, r= 0.29; P=0.03). PVN IL-6 content was also correlated with HR variability (Y=-1.8x+21.5, r=-0.36; P=0.01), BrS (Y= -0.3x+4.2, r=-0.44; P<0.001) and HR (Y=7x+324, r=0.30; P=0.02). Conclusions: Data suggested the involvement of PVN pro-inflammatory cytokines in the mediation of cardiovascular deficits in the SHR and the efficacy of T to prompt normalize these deleterious effects.


Sign in / Sign up

Export Citation Format

Share Document