Hypertension and Brain Inflammation: Role of RAS-Induced Glial Activation



Fig. 9.1
Hypothesis for PRO/PRR and Ang II/AT1R interactions in pre-autonomic nuclei during neurogenic hypertension. RAS-induced astrogliosis increases pro-inflammatory cytokine expression and microglial recruitment at pre-autonomic nuclei such as the PVN, thus enhancing neuroimmune communication and resulting in neurogenic hypertension. PRO Prorenin, PRR Prorenin Receptor, Ang II Angiotensin II, AT1R Angiotensin II Type 1 Receptor, IL-1β Interleukin 1β, IL-6 Interleukin 6, TNF-α Tumor Necrosis Alpha, MCP-1 Monocyte Chemoattractant Protein 1



Microglial-derived cytokine release may also act in an auto- and paracrine fashion activating neighboring cells such as astrocytes, which in similar fashion to microglia, facilitate the synthesis and secretion of a variety of inflammatory cytokines and chemokines (Fig. 9.1). In the brain, astrocytes are considered the primary source of MCP-1 production, the main role of which is to attract and recruit monocytes and macrophages into inflammatory sites [6971]. Thus, MCP-1 production by astrocytes at the PVN helps create a niche of inflammation at the nucleus. In turn, activated microglia also release glutamate while simultaneously disrupting the efficiency of glutamate uptake by astrocytes [72]. Excess of glutamate concentration at the synaptic cleft results in enhanced neuronal depolarization and excitability thus increasing the PVN-RVLM-IML communication, releasing vasopressin, increasing norepinephrine spillover, and enhancing sympathetic outflow [8, 15]. As a result, sympathetic input to organs such as the heart, vasculature, and kidney is increased, thus contributing to ventricular hypertrophy, vasoconstriction, and renin release. Coupled to this is the enhancement of the peripheral inflammation through sympathetic input to the gut and spleen, which are two major regulatory players in the peripheral immune response. Hence, coming full circle and perpetuating the RAS-Immune-CNS interactions that leads to blood pressure increase.




6 Conclusions


Unlike drastic scenarios such as head trauma or stroke where an intracerebral pro-inflammatory response causes cell death, evidence indicates that the development of hypertension which is neurogenic in origin (chronic sympathoexcitation) involves a milder and persistent increase in intrinsic pro-inflammatory processes within brain cardiovascular control centers. In concert with this, it is also apparent that systemic pro-inflammatory responses that result from chronic hypertension can influence brain cardiovascular control centers, including significant activation of intracerebral pro-inflammatory mechanisms that ultimately affect blood pressure. Nonetheless, despite much progress towards a better understanding the pathophysiological processes in hypertension, many other questions remain to be answered. First and foremost is which of the above described scenarios comes first? For example, is the initiating factor high blood pressure that in turn activates, via systemic and intracerebral inflammation, mechanisms that result in chronic sympathoexcitation? Or, is the “chicken” the co-activation via the RAS of pre-autonomic and cerebral immune mechanisms that elicit chronic sympathoexcitation, and the “egg” the resulting hypertension and systemic immune activation that then feeds back to the brain to amplify the pro-immune cycle. Other questions remain, and include: (1) What are the contributions of brain cardiovascular control centers other than the PVN to the neuroinflammatory processes that contribute to hypertension? Is neuroinflammation in hypertension something that is peculiar to the PVN, or is it a generalized phenomenon for all brain centers that play a role in cardiovascular regulation. (2) How can the challenge of delivering drugs to the brain cardiovascular control centers across an impermeable BBB be solved, in order to design better treatment options for drug-resistant hypertensive patients? Or is this even necessary, as it has been shown that the BBB becomes leaky in chronic hypertension [27]?

One thing has become clear during the past few years, namely that enhanced neuroinflammation and overactivation of the RAS are major contributors to neurogenic hypertension. Hence, identifying the basic mechanisms involved in RAS-induced inflammation at cardioregulatory regions in the brain serves as a good starting point to further characterize the mechanisms underlying the onset and progress of this disease. To this end, while the majority of studies have focused on neuronal actions of Ang II/AT1R activation within the CNS on blood pressure control and hypertension, it is now well recognized that both Ang II and PRO can influence, directly or indirectly, the activity and immune function of both astrocytes and microglia [19, 31, 64]. Furthermore, as research indicates that controlling for inflammation is also favorable for hypertensive patients, should anti-inflammatory drugs be considered as a first line of defense in treating this condition? We present evidence that both microglia and astrocytes play a significant role in this disease thus making these cells a new therapeutic target when treating hypertension. Interestingly, despite the similarities between microglial and astrocyte activation, the contribution of astrocytes during RAS-induced hypertension, and to neuroinflammatory process and consequent blood pressure regulation during neurogenic hypertension has not been fully investigated.


References



1.

Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, et al. Heart disease and stroke statistics—2014 update: a report from the American Heart Association. Circulation. 2014;129(3):e28–292.PubMedCrossRef


2.

Li JJ, Fang CH, Hui RT. Is hypertension an inflammatory disease? Med Hypotheses. 2005;64(2):236–40.PubMedCrossRef


3.

Boos CJ, Lip GY. Is hypertension an inflammatory process? Curr Pharm Des. 2006;12(13):1623–35.PubMedCrossRef


4.

Lazartigues E. Inflammation and neurogenic hypertension: a new role for the circumventricular organs? Circ Res. 2010;107(2):166–7.PubMedPubMedCentralCrossRef


5.

Daniels D, Mietlicki EG, Nowak EL, Fluharty SJ. Angiotensin II stimulates water and NaCl intake through separate cell signalling pathways in rats. Exp Physiol. 2009;94(1):130–7.PubMedPubMedCentralCrossRef


6.

Fyhrquist F, Metsarinne K, Tikkanen I. Role of angiotensin II in blood pressure regulation and in the pathophysiology of cardiovascular disorders. J Hum Hypertens. 1995;9 Suppl 5:S19–24.PubMed


7.

Santos RA. Angiotensin-(1-7). Hypertension. 2014;63(6):1138–47.PubMedCrossRef


8.

Nunes FC, Braga VA. Chronic angiotensin II infusion modulates angiotensin II type I receptor expression in the subfornical organ and the rostral ventrolateral medulla in hypertensive rats. J Renin Angiotensin Aldosterone Syst. 2011;12(4):440–5.PubMedCrossRef


9.

Marvar PJ, Thabet SR, Guzik TJ, Lob HE, McCann LA, Weyand C, et al. Central and peripheral mechanisms of T-lymphocyte activation and vascular inflammation produced by angiotensin II-induced hypertension. Circ Res. 2010;107(2):263–70.PubMedPubMedCentralCrossRef


10.

Elenkov IJ, Chrousos GP. Stress hormones, proinflammatory and antiinflammatory cytokines, and autoimmunity. Ann N Y Acad Sci. 2002;966:290–303.PubMedCrossRef


11.

Li DP, Pan HL. Glutamatergic inputs in the hypothalamic paraventricular nucleus maintain sympathetic vasomotor tone in hypertension. Hypertension. 2007;49(4):916–25.PubMedCrossRef


12.

Badoer E. Hypothalamic paraventricular nucleus and cardiovascular regulation. Clin Exp Pharmacol Physiol. 2001;28(1–2):95–9.PubMedCrossRef


13.

Li YF, Jackson KL, Stern JE, Rabeler B, Patel KP. Interaction between glutamate and GABA systems in the integration of sympathetic outflow by the paraventricular nucleus of the hypothalamus. Am J Physiol Heart Circ Physiol. 2006;291(6):H2847–56.PubMedCrossRef


14.

Affleck VS, Coote JH, Pyner S. The projection and synaptic organisation of NTS afferent connections with presympathetic neurons, GABA and nNOS neurons in the paraventricular nucleus of the hypothalamus. Neuroscience. 2012;219:48–61.PubMedPubMedCentralCrossRef


15.

Braga VA, Medeiros IA, Ribeiro TP, Franca-Silva MS, Botelho-Ono MS, Guimaraes DD. Angiotensin-II-induced reactive oxygen species along the SFO-PVN-RVLM pathway: implications in neurogenic hypertension. Braz J Med Biol Res. 2011;44(9):871–6.PubMedCrossRef


16.

Grobe JL, Xu D, Sigmund CD. An intracellular renin-angiotensin system in neurons: fact, hypothesis, or fantasy. Physiology (Bethesda). 2008;23:187–93.CrossRef


17.

Nguyen G, Contrepas A. Physiology and pharmacology of the (pro)renin receptor. Curr Opin Pharmacol. 2008;8(2):127–32.PubMedCrossRef


18.

Nguyen G, Delarue F, Burckle C, Bouzhir L, Giller T, Sraer JD. Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin. J Clin Invest. 2002;109(11):1417–27.PubMedPubMedCentralCrossRef

Oct 29, 2016 | Posted by in NEUROSURGERY | Comments Off on Hypertension and Brain Inflammation: Role of RAS-Induced Glial Activation

Full access? Get Clinical Tree

Get Clinical Tree app for offline access