Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jun 1.
Published in final edited form as: Hypertension. 2011 May 2;57(6):1026–1033. doi: 10.1161/HYPERTENSIONAHA.111.169748

Autonomic-Immune-Vascular Interaction: An Emerging Concept for Neurogenic Hypertension

Jasenka Zubcevic #, Hidefumi Waki ¥, Mohan K Raizada #,, Julian FR Paton *,
PMCID: PMC3105900  NIHMSID: NIHMS295117  PMID: 21536990

INTRODUCTION

Hypertension is the single most important risk factor for cardiovascular disease and thus remains a global public health challenge. Significant progress has been made during the last several decades in the treatment of hypertension through the use of inhibitors of the renin-angiotensin system (RAS), with either angiotensin converting enzyme inhibitors (ACEIs) or angiotensin receptor type 1 (AT1R) blockers (ARB), diuretics, α-adrenoreceptor antagonists, and calcium channel blockers. Despite these advances, it has been extremely difficult to manage hypertension in ~40% of hypertensive patients1. A majority of these unresponsive patients exhibit increased sympathetic drive and display neurogenic components2,3,4. The role of elevated sympathetic nervous system in drug-resistant hypertension is corroborated by the recent successes in its treatment by renal sympathetic denervation in humans5,6,7. These observations have led to the realization that elucidation and understanding of cellular, molecular, and physiological mechanisms involved in the development and establishment of neurogenic hypertension is pivotal in advancing new therapeutic strategies which would be effective in a majority of patients. The objective of this article is to summarize recent advances in this field, underscoring the importance of coordinated neural-peripheral signaling in neural control of blood pressure (BP), contribution of inflammatory molecules, and to introduce novel and emerging concepts in the pathogenesis of neurogenic hypertension. It is advised to consult other reviewers for details on fundamental studies related to neural pathways, renin-angiotensin system (RAS), and involvement of other hormonal systems in neurogenic hypertension8,9,10,11,12,13,14.

Neurogenic hypertension

An alteration in neural cardiovascular (vagal/sympathetic) control mechanism is the principal characteristic of neurogenic hypertension. Studies demonstrating a significant enhancement of the vasomotor and cardiac sympathetic drive, as well as a reduction in the parasympathetic drive in borderline hypertensive patients, support this premise15,16,17. In addition, hypertensive patients show a greater plasma norepinephrine (NE) level, and greater NE spillover in both the peripheral circulation and blood draining from the brain as well as raised sympathetic postganglionic activity targeting the skeletal muscle vascular bed18,19,20,21. Animal models corroborate the human data. Recent evidence shows that even the monogenic forms of hypertension, such as the apparent mineralocorticoid excess, which greatly depends on salt absorptive mechanisms of the kidney, have also been attributed to an elevated sympathetic drive indicating a very close association between renal dysfunction and sympathetic overdrive22,23. Equally, the Goldblatt model partially depends on signaling within the rostral ventrolateral medulla (RVLM) for the maintenance of hypertension24.

Evidence from the spontaneously hypertensive rat (SHR), a well-established animal model for human hypertension, supports the idea that sympathetic overdrive precedes hypertension25. Therefore, molecular and neuronal changes at the level of the brainstem and hypothalamus may contribute to neurogenic hypertension. Our studies have demonstrated that elevated RAS in the brainstem and hypothalamus of juvenile SHR affects the regulation of arterial pressure and baroreceptor reflex gain in the adult SHR26,27,28,29. Moreover, enhanced central respiratory-sympathetic coupling in rat models, of which are already present at a pre-hypertensive age, contributes to raise vasomotor tone25,30,31,32. Thus, sympathetic activation appears to not only initiate hypertension, but also maintains it. Whether elevated sympathetic activity is causal to the initiation of hypertension remains equivocal, but it certainly occurs early in the condition.

Inflammation, the blood brain barrier and hypertension

Overwhelming evidence, both clinical and experimental, demonstrates that hypertension is associated with circulating levels of inflammation markers. In particular, there is strong support of the concept that the vascular inflammatory process plays an important role in the pathophysiology of hypertension (Figure 1). Cross-sectional and prospective studies have demonstrated that circulating levels of inflammatory molecules such as c-reactive protein (CRP), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and adhesion molecules such as P-selectin and intercellular adhesion molecule-1 (ICAM-1), are increased in patients with primary hypertension33,34,35,36,37. Women's Health Study indicated that CRP levels predicted the development of hypertension, an association that was independent of BP. Finally, inhibitors of the RAS, which are effective anti-hypertensive in some patients, are shown to decrease CRP, IL-6, TNF- α levels, and inflammatory status33,34,35,36,37. Animal studies have supported these clinical association studies and have been instrumental in furthering our understanding of the involvement of inflammatory mechanisms in vascular dysfunction and the hypertensive condition. For example, total leukocyte counts in young, mature, and old SHR were 50-100% above the controls with the number of monocytes, activated monocytes, and the lymphocyte counts significantly elevated38. Leukocyte-endothelial interactions were also reported to be altered in the SHR38. Moreover, the elevated BP, above normal leukocyte counts, and elevated number of activated neutrophils in SHR, were all suppressed after adrenalectomy, indicating a relationship between altered leukocyte adhesiveness and adrenal corticosteroids in SHR38. Blood transfusions from SHR to Wistar-Kyoto (WKY) rats increased BP due to the more sticky nature of the SHR leukocytes39. The ‘stickier’ nature of leukocytes has been attributed to the elevated glucocorticoid levels in the SHR, which induces leukocyte pseudopod projections and results in slower cell passage through capillaries and elevated hemodynamic resistance in the SHR39. Furthermore, elevated Ang II promotes leukocyte-endothelial interaction, thereby contributing to vascular inflammation40. T cells play an important role in Ang II infusion-, DOCA-salt-induced hypertension, and associated vascular and renal dysfunctions41,42,43. T cell modulating agent, mycophenolate mofetil, prevents hypertension in animal models43. The SHR exhibits increased levels of activated monocytes whose levels are significantly decreased by reducing BP38. Vascular inflammation in this rat model of hypertension was increased along with increases in expression of inflammatory cytokines interleukin-1β (IL-1β), IL-6, and TNF-α. AT1 receptor blocker, Candesartan, produced anti-hypertensive effects and decreases in inflammatory cytokines34,44. Similarly, an association between inflammation and hypertension has been demonstrated in aldosterone models. For example, circulating levels of inflammatory markers are increased by aldosterone infusion in humans and patients with primary hyperaldosteronism, which is associated with vascular dysfunction, cardiac fibrosis, and increased risk of myocardial infarction and stroke33. Chronic treatment of hypertensive rats with mineralocorticord receptor antagonist, which attenuates BP, decreases circulating levels of certain inflammatory cytokines, and renal and cardiac expression of inflammatory mediators such as IL-1, IL-6 and nuclear factor kappa beta (NFκβ)33.

Figure 1. Proposed hypothesis of a dysfunctional autonomic-immune-vascular mechanism in neurogenic hypertension.

Figure 1

A coordinated peripheral-brain communication is critical in the development and establishment of neurogenic hypertension. AT1 receptors on SFO/OVLT and other CVOs are activated by increased plasma Ang II and an integrated signal is transmitted to the PVN. This results in the activation of microglia and increased generation of ROS and cytokines and decrease in NO. This leads to increased neuronal activity by regulation of ion channel function directly or indirectly. Ang II generated within the PVN can also activate neuronal AT1 receptors to compliment changes in neuronal activity. An imbalance in sympathetic/parasympathetic activities can directly influence bone marrow activity resulting in increase in pro-inflammatory cells (PICs, increase inflammation) and decrease in EPCs (decrease in vascular repair). All these events would lead to high BP and associated CV pathophysiology. These neural-vascular signals are further perpetuated by AT1 receptor-mediated changes in cerebral blood vessels, interrupting the integrity of BBB and allowing inflammatory cells to enter the brain parenchyma, contributing to microglial activation and inflammation in the PVN. A similar AT1-dependent mechanism may be present in other cardioregulatory brain regions such as the NTS and the RVLM.

The association between inflammation and inflammatory cytokines and chemokines and hypertension is evident from the above discussion. However, the concept of the involvement of the brain in inflammatory mechanisms is still evolving. For example, inflammatory cytokines such as TNF-α and IL-1β induce cyclooxygenase-2 (COX-2) activity in perivascular macrophages of the blood brain barrier (BBB) and generate prostaglandin E2, which enters the brain and stimulates paraventricular nucleus (PVN) neurons regulating adrenocorticotropic hormone (ACTH) release and sympathetic drive45,46. Additionally, expression of junctional adhesion molecule-1 (JAM-1) is significantly increased in the nucleus of the solitary tract (NTS) of SHR relative to WKY rats47. This increased expression of JAM-1 induces both hypertension (in normotensive rats) and leukocytes accumulation in the NTS microvasculature and may lead to enhancement of cell transmigration across the tight junctions, activation of platelet aggregation, and increased production of cytokines47, thereby contributing to the inflammatory status of the NTS which may then directly or indirectly regulate NTS neuronal activity and baroreceptor functions. Saavedra and collaborators have also demonstrated that inflammation in the SHR is not restricted to the peripheral vessels but it is also extended to the cerebral vasculature44. Treatment of SHR with angiotensin receptor blockers with access to the brain, reversed cerebrovascular inflammation by decreasing macrophage infiltration, normalization of eNOS/iNOS ratio, reversal of heat-shock protein up regulation, and reversal of increased TNF-α, IL-1β, and ICAM44. These observations beg the question: are inflammatory conditions in the cardiovascular control regions of the brain a result of cytokines produced in the circulation and exerting their influence in the cerebral vasculature to initiate a cascade of events leading to increased neuronal activity and sympathetic tone? Alternatively, are cytokines produced within the brain to regulate neuronal activity? There is no conclusive answer to support or refute either contention at the present time. However, multiple studies support the latter view. They include the following: (i) increased expression of inflammatory cytokines has been demonstrated in cardiovascular relevant brain regions of various animal models of hypertension48,49,50,51 and in the heart failure model45,52; (ii) ICV infusion of inflammatory cytokines such as IL-1α activates the sympathetic nervous system and increases BP53,54; (iii) overexpression of IL-10 in the CNS following ICV administration decreases TNF-α, IL-1α, PGE2, and COX-2 in the PVN and ameliorates sympathoexcitation55; (iv) AAV-mediated neuronal expression of IL-10 in the PVN attenuates chronic Ang II-induced hypertension51; (v) IL-6 in the NTS depresses the baroreceptor reflex gain56; (vi) Migratory inhibitory factor (MIF), an anti-inflammatory cytokine, has been shown to negatively regulate chronotropic actions of angiotensin II in the neurons57. Thus, decreased MIF expression in the PVN in response to angiotensin II may be associated with the development of high BP in the SHR50; and (vii) numerous studies have demonstrated that inflammatory cytokines have profound effects on neuronal activity directly. For example, TNF-α and IL-1α increases neuronal firing frequency by binding to their respective receptors and the activation of NFkβ58. Consistent with this, is the observation that ICV infusion of a specific NFkβ inhibitor (pyrrolidine dithiocarbamate) significantly attenuates Ang II-induced hypertension and inflammatory cytokines in the PVN49. Cytokines can modulate neuronal activity via production of reactive oxygen species (ROS) and activation of iNOS. Resulting nitric oxide (NO) diffuses to adjacent tissues to modulate neuronal activity by influencing various ion channels59,60. (viii) Receptors for cytokines are found in several sites in the brain on multiple cell types including neurons, microglia and astrocytes61. There is the possibility that a combination of both mechanisms (i.e. contribution of peripheral cytokines via cerebral vasculature and generation of intrinsic cytokines within the brain) may contribute to the neuroinflammatory process in neurogenic hypertension. It appears that the inflammatory process in the cardiovascular regulatory regions of the brain is linked with modulation of the autonomic nervous system and the increased BP. However, we acknowledge that not all inflammation in the brain results in hypertension and propose unique inflammatory profiles relate to distinct disease states48. Finally, animal and human studies indicate hypoperfusion of the hypertensive brain can occur due to increased cerebral vascular resistance62,63. The relationship between hypoperfusion and inflammation within brain microvasculature is undetermined but one can trigger the other thereby presenting a possible mechanism.

Brain microglia, cytokines, and neurogenic hypertension

Microglia, the resident macrophages of the brain, have attracted significant attention in recent years as a result of their role in various neuropathological conditions. They are activated rapidly in response to brain injury and produce a variety of inflammatory mediators including cytokines. In ischemic stroke, for example, microglia exerts neurotoxic functions through production of ROS and cytokines. This damages the BBB integrity leading to infiltration of inflammatory cells into the brain64,65. Activated microglial cells are present in the CNS from patients with chronic neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis66,67. Their importance in these disease processes has been studied extensively.

The concept that microglial cells in the cardiovascular relevant brain regions may participate in hypertension-linked pathophysiology is derived, in part, from our studies which show that chronic Ang II-induced hypertension is associated with increases in activated microglia in the PVN51. This activation is associated with increases in the levels of PVN inflammatory cytokines. Similarly, we found an increase in activated microglia in the PVN of the SHR (Figure 2). Further evidence in support of this concept is provided by our experiments with minocycline, an antibiotic which can cross BBB and inhibit microglial activation51. ICV infusion of minocycline attenuates Ang II-induced high BP, decreases PVN cytokines, and attenuates cardiac hypertrophy51. Multiple examples in the literature indirectly support the role of brain microglia. Saavedra et al have demonstrated that acute brain inflammatory response to bacterial endotoxin lipopolysaccharide results in the activation of microglia, not only in the cardiovascular-relevant brain regions (PVN, SFO) but also in prefrontal cortex, hippocampus, and amygdala68,69. This is associated with increases in brain expression of inflammatory cytokines and their receptors, adhesion molecules, and iNOS68,69. Systemic administration of centrally acting AT1 receptor blockers decreases these responses. Carnevale et al studied interactions among hypertension, inflammation, and β-amyloid (Aβ) deposition in a hypertensive mouse with transverse aortic coarctation70. They demonstrated that hypertension, per se, triggers microglia activation and neuroinflammation before the Aβ deposition takes place, suggesting that microglial activation precedes and may trigger neurodegenerative processes in Alzheimer's disease. Finally, Lanz et al provided evidence that Ang II sustains brain inflammation in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis via the transforming growth factor β (TGF-β)71. Their studies indicate that neurons and microglial cells express AT1 receptors which are primarily responsible for Ang II-induced increases in TGF-β production. Collectively, these observations provide persuasive arguments in support of brain for transmitting inflammatory signals that spread across BBB into cardiovascular control regions of the brain.

Figure 2. Activated microglial cells in the PVN of WKY rat and SHR.

Figure 2

Immunostaining with OX-42 antibody was used to identify activated microglia as described elsewhere51. Quantitation based on morphological analysis revealed a 75% increase in activated microglial cells in the PVN of SHR vs. WKY rat.

Visceral control of inflammation and putative alterations in neurogenic hypertension

The brain is devoid of the “classical immuno-surveillance system” but is in constant vigilance of the inflammatory status of the body by a reflex afferent feedback system. In recent years, the concept of “vagal immunoflex” has been introduced. This neuronal circuit involves activation of the vagal nerve afferents sensitive to systemic inflammation and reflex release of acetylcholine (Ach) in organs such as the liver, spleen, bone marrow, and heart to dampen the tissue inflammation by reducing pro-inflammatory cytokines. Presence of specific Ach receptors (nAchRα 7) on inflammatory cells support this anti-inflammatory role72,73,74. In addition, vagal afferents can directly sense cytokine levels in the blood via specific cytokine receptors such as IL-1R present on the glomus cells adjacent to vagus nerve afferent endings within paraganglia located within the gastrointestinal tract, spleen, bone marrow, liver, and heart75,76,77,78,79. Thus, vagal immunoflex system presumably transmits signals to the NTS, the central site of vagal termination, before onward relay to dorsal vagal motoneurons.

It is concluded from the above discussion that the vagus nerve is vital in informing the brain of the peripheral immune status and controlling the activity of the immune cells. However, is this effective in hypertension? In hypertension, cardiac vagal tone and the cardiac vagal baroreflex gain are both depressed80,81. Similarly, we propose that the anti-inflammatory function of the vagus is also suppressed in conditions of hypertension. The possibility that the raised levels of sympathetic activity (vasomotor nerves) associated with hypertension is also pro-inflammatory, is a hypothesis at this point.

Dysfunctional ANS, endothelial repair, and neurogenic hypertension

Could dysfunctional cardiovascular autonomic activity in neurogenic hypertension suppress endothelial repair? It is well-accepted that endothelial dysfunction is an early event in hypertension-induced vascular pathophysiology. The healthy endothelial cells lining the blood vessels have both mechanical and functional roles, producing NO to promote vasodilation and reducing oxidative stress and inflammation. Since mature endothelial cells have a limited regenerative capacity, endothelial progenitor cells (EPCs) contribute to the repair and maintenance of endothelial damage to maintain normal vascular homeostasis in healthy individuals. In hypertension, however, dysfunctional endothelial cells produce ROS and other inflammatory molecules, which result in vasoconstriction, platelet activation, inflammation, and fibrosis82,83,84,85. In addition, EPC numbers, migratory ability and functions are all impaired in hypertension86. A combination of these processes leads to accelerated vascular dysfunction and hypertension-associated pathophysiology; therefore, (i) are EPCs from hypertensives dysfunctional, and, (ii) is there a neural control mechanism for EPC release and function that are altered in neurogenic hypertension?

  1. EPC numbers and function have been inversely correlated in patients with cardiovascular disease (CVD), obesity, diabetes, chronic kidney disease, and other immune diseases87,88,89,90. Similarly, decreases in numbers as well as their functions have been demonstrated in human hypertensives and animal models of hypertension91,83,84,86. Dysfunctional EPCs are associated with increases in EPCs ROS, NADPH oxidase, and decreases in NO production. This view is supported by evidence that antihypertensive drugs such as ACE inhibitors and ARBs increase EPCs numbers and improve their function92,93.

  2. Release and function of EPCs from bone marrow appears to be neurally-regulated. Bone marrow is densely innervated by the sympathetic nervous system which stimulates the release of bone marrow-derived stem cells into the circulation94,95,96,97,98. Similarly, cholinergic fibers have been found in the rat femur bone marrow around hematopoietic cells, suggesting parasympathetic regulation bone marrow-derived cells as well98. Finally, retrograde tracing studies with rabies virus have clearly indicated a connection between the bone marrow and many cardiovascular control regions of the brain99.

Considered together, these data suggest that dysfunctional sympathetic/parasympathetic regulation of bone marrow could lead to impaired EPCs levels and their repair capacity in hypertension. An altered sympathetic (and/or parasympathetic) drive to the bone marrow may contribute to EPC dysfunction in hypertension. These proposals are supported by the following evidence: (i) Decrease in sympathetic activity to the bone marrow in diabetes is associated with decreases in circulating EPCs which are also dysfunctional97,88, and (ii) while it is generally considered that sympathetic activity is increased in hypertension, separate end organs can be differentially regulated by the sympathetic nervous system, and certain vascular beds reportedly receive decreased sympathetic activity in hypertension100,101. It is tempting to suggest that a decreased sympathetic drive to the bone marrow could contribute to its dysfunction. Additionally, a decreased parasympathetic (vagal) regulation of the bone marrow may instead contribute in its dysfunction (Figure 1).

Unifying hypothesis

Synthesis of all available evidence has led us to propose the following hypothesis as a mechanism for neurogenic hypertension (Figure 1). Increase in peripheral Ang II leads to a cascade of events: AT1 receptors in the neurons of CVOs are activated resulting in increases in ROS102,13,103. This leads to the activation of microglia in the PVN (Figure 2) which increases production of inflammatory cytokines, ROS, and other inflammatory modulators51. These modulators would stimulate PVN neuronal activity which, via brainstem (NTS and RVLM), would be reflected in modulation of ANS leading to increases in blood pressure. An imbalance in sympathetic/parasympathetic activities also regulates bone marrow activity by increasing inflammatory cells and decreasing EPCs and their functions. Decreases in EPC numbers and their dysfunctions accelerate vascular dysfunction and damage. Increases in inflammatory cells would result in increased inflammatory mediators which would exaggerate vascular dysfunction leading to the development of pathophysiology of cardiovascular-relevant tissue characteristics of hypertension, as well as issues of hypoperfusion63.

The vascular arm of the neurovascular communication involves activation of AT1 receptors on the cerebral vessels which have free access to increased levels of peripheral Ang II. Activation of these receptors stimulates cytokines, JAM-1, ROS, and adhesion molecules. This interrupts the integrity of the BBB104,105,106,107 and enhances adherence of inflammatory cells causing leukocytes and cytokines to enter the brain parenchyma, contributing to microglial activation and inflammatory status of the PVN. Similar process could occur in other cardioregulatory brain regions.

Future directions

Although some evidence, as presented in this review, exists in overall support of the above hypothesis, many important issues remain to be proven. Further evidence would be needed in order to provide conclusive validation for our hypothesis.

  1. In our hypothesis we have proposed a central role for the PVN. This is primarily based on our data that shows activated microglia predominantly in this region (51, Figure 2). However, the simultaneous involvement of a similar mechanism in other brain regions (i.e. NTS, RVLM, etc.) cannot be ruled out and needs to be investigated.

  2. The hypothesis proposes that increases in activated microglia in the brain are both resident and derived from the peripheral inflammatory cells. There is no evidence to support this in hypertension. However, recent evidence from Chen et al108 has shown that defective microglia cause pathological grooming in Hoxb8 mutant mice. Bone marrow transplant from wild type mice increases brain microglia and cures Hoxb8 mice of pathological grooming, indicating that activated microglia in this animal model result from immune cells in the peripheral circulation. Additionally, Longo et al109 has demonstrated that a significant number of activated microglia observed in the hippocampus of status epileptus mice have bone marrow origin. Similar transplantation studies are needed to determine the origin of activated microglia in the brain in hypertension.

  3. Are microglia targets of Ang II actions? It is without dispute that neurons in the cardiovascular regulatory brain regions such as the PVN express AT1 receptors and Ang II interaction with these receptors profoundly affects neuronal activity by stimulating a cascade of signaling pathways110,111. It remains to be proven, however, whether AT1 receptors are present on microglia and if so, how they contribute to increased neuronal activity in neurogenic hypertension. Recent studies have shown a co-localization of AT1 receptor with Iba-1 positive cells, a marker for microglia in the brain of mouse model of multiple sclerosis71. In addition, TGF-β is co-localized with AT1 receptors in microglia. Furthermore, Lanz TV et al71 showed that EAE-induced TGF-β expression is blocked by Candesartan, an AT1 receptor blocker concluding that microglia contains functional AT1 receptors. Evidence for this conclusion is provided by in vitro studies which demonstrate that microglial cells in culture increase TGF-β expression in response to Ang II71. How these receptors regulate generation of inflammatory cytokines, ROS, and other mediators to influence neuronal activity, remains to be investigated.

  4. We have hypothesized that a functional balance between the inflammatory cells and EPCs from the bone marrow is important in the maintenance of normal cardiovascular physiology. Its imbalance leads to vascular pathophysiology associated with hypertension. This concept needs to be further investigated.

Perspectives

Despite recent advances, it has been extremely difficult to successfully manage and control hypertension in a significant number of hypertensive patients. A majority of these unresponsive patients exhibit increased sympathetic drive and display neurogenic hypertension. Lack of underlying mechanisms involving neural-peripheral communication associated with increased sympathetic activity has contributed to this. We have proposed a unifying hypothesis of a dysfunctional neural-immune-vascular communication pathway that may be responsible for neurogenic hypertension. Evidence to support/refute this hypothesis will be critical in the elucidation of the underlying impairment, and exploring the mechanism for the advancement of neurogenic hypertension therapeutics.

Acknowledgments

SOURCES OF FUNDING This work was supported by the NIH (R37 HL33610) and British Heart Foundation (RG/07/006/23634). JFRP was in receipt of a Royal Society Wolfson Research Merit Award.

Footnotes

DISCLOSURES None.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Primatesta P, Brookes M, Poulter NR. Improved hypertension management and control: results from the health survey for England 1998. Hypertension. 2001;38:827–832. [PubMed] [Google Scholar]
  • 2.Mann SJ. Neurogenic essential hypertension revisited: the case for increased clinical and research attention. Am J Hypertens. 2003;16:881–888. doi: 10.1016/s0895-7061(03)00978-6. [DOI] [PubMed] [Google Scholar]
  • 3.Grassi G, Quarti-Trevano F, Brambilla G, Seravalle G. Blood pressure control in resistant hypertension: new therapeutic options. Expert Rev Cardiovasc Ther. 2010;8:1579–1585. doi: 10.1586/erc.10.138. [DOI] [PubMed] [Google Scholar]
  • 4.Fisher JP, Fadel PJ. Therapeutic strategies for targeting excessive central sympathetic activation in human hypertension. Exp Physiol. 2010;95:572–580. doi: 10.1113/expphysiol.2009.047332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Myrvang H. Hypertension: the simplicity of renal denervation for patients with treatment-refractory hypertension. Nat Rev Nephrol. 2011;7:64. doi: 10.1038/nrneph.2010.181. [DOI] [PubMed] [Google Scholar]
  • 6.Mearns BM. Hypertension: is renal denervation a cure for drug-resistant disease? Nat Rev Cardiol. 2011;8:2. doi: 10.1038/nrcardio.2010.190. [DOI] [PubMed] [Google Scholar]
  • 7.Grassi G, Quarti-Trevano F, Brambilla G, Seravalle G. Blood pressure control in resistant hypertension: new therapeutic options. Expert Rev Cardiovasc Ther. 2010;8:1579–1585. doi: 10.1586/erc.10.138. [DOI] [PubMed] [Google Scholar]
  • 8.Cuadra AE, Shan Z, Sumners C, Raizada MK. A current view of brain reninangiotensin system: Is the (pro)renin receptor the missing link? Pharmacol Ther. 2010;125:27–38. doi: 10.1016/j.pharmthera.2009.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ferreira AJ, Santos RA, Bradford CN, Mecca AP, Sumners C, Katovich MJ, Raizada MK. Therapeutic implications of the vasoprotective axis of the reninangiotensin system in cardiovascular diseases. Hypertension. 2010;55:207–213. doi: 10.1161/HYPERTENSIONAHA.109.140145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Xu P, Sriramula S, Lazartigues E. ACE2/Ang-(1-7)/Mas pathway in the brain: The Axis of Good. Am J Physiol Regul Integr Comp Physiol. 2010;300:R804–R817. doi: 10.1152/ajpregu.00222.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Grassi G, Seravalle G, Quarti-Trevano F. The ‘neuroadrenergic hypothesis’ in hypertension: current evidence. Exp Physiol. 2010;95:581–586. doi: 10.1113/expphysiol.2009.047381. [DOI] [PubMed] [Google Scholar]
  • 12.Grassi G, Quarti-Trevano F, Dell'oro R, Mancia G. Essential hypertension and the sympathetic nervous system. Neurol Sci. 2008;29(Suppl 1):S33–S36. doi: 10.1007/s10072-008-0882-9. [DOI] [PubMed] [Google Scholar]
  • 13.Osborn JW, Fink GD, Sved AF, Toney GM, Raizada MK. Circulating angiotensin II and dietary salt: converging signals for neurogenic hypertension. Curr Hypertens Rep. 2007;9:228–235. doi: 10.1007/s11906-007-0041-3. [DOI] [PubMed] [Google Scholar]
  • 14.Zoccal DB, Paton JF, Machado BH. Do changes in the coupling between respiratory and sympathetic activities contribute to neurogenic hypertension? Clin Exp Pharmacol Physiol. 2009;36:1188–1196. doi: 10.1111/j.1440-1681.2009.05202.x. [DOI] [PubMed] [Google Scholar]
  • 15.Grassi G, Bombelli M, Seravalle G, Dell'Oro R, Quarti-Trevano F. Diurnal blood pressure variation and sympathetic activity. Hypertens Res. 2010;33:381–385. doi: 10.1038/hr.2010.26. [DOI] [PubMed] [Google Scholar]
  • 16.Grassi G, Seravalle G, Trevano FQ, Dell'oro R, Bolla G, Cuspidi C, Arenare F, Mancia G. Neurogenic abnormalities in masked hypertension. Hypertension. 2007;50:537–542. doi: 10.1161/HYPERTENSIONAHA.107.092528. [DOI] [PubMed] [Google Scholar]
  • 17.Grassi G. Assessment of sympathetic cardiovascular drive in human hypertension: achievements and perspectives. Hypertension. 2009;54:690–697. doi: 10.1161/HYPERTENSIONAHA.108.119883. [DOI] [PubMed] [Google Scholar]
  • 18.Anderson EA, Sinkey CA, Lawton WJ, Mark AL. Elevated sympathetic nerve activity in borderline hypertensive humans: Evidence from direct intraneural recordings. Hypertension. 1989;14:177–183. doi: 10.1161/01.hyp.14.2.177. [DOI] [PubMed] [Google Scholar]
  • 19.Grassi G. Role of the sympathetic nervous system in human hypertension. J Hypertens. 1998;16:1979–1987. doi: 10.1097/00004872-199816121-00019. [DOI] [PubMed] [Google Scholar]
  • 20.Esler M, Jennings G, Lambert G. Noradrenaline release and the pathophysiology of primary human hypertension. Am J Hypertens. 1989;2:140S–146S. doi: 10.1093/ajh/2.3.140s. [DOI] [PubMed] [Google Scholar]
  • 21.Ferrier C, Esler MD, Eisenhofer G, Wallin BG, Horne M, Cox HS, Lambert G, Jennings GL. Increased norepinephrine spillover into the jugular veins in essential hypertension. Hypertension. 1992;19:62–69. doi: 10.1161/01.hyp.19.1.62. [DOI] [PubMed] [Google Scholar]
  • 22.Bailey MA, Paterson JM, Hadoke PW, Wrobel N, Bellamy CO, Brownstein DG, Seckl JR, Mullins JJ. A switch in the mechanism of hypertension in the syndrome of apparent mineralocorticoid excess. J Am Soc Nephrol. 2008;19:47–58. doi: 10.1681/ASN.2007040401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Krum H, Schlaich M, Whitbourn R, Sobotka PA, Sadowski J, Bartus K, Kapelak B, Walton A, Sievert H, Thambar S, Abraham WT, Esler M. Catheter-based renal sympathetic denervation for resistant hypertension: a multicentre safety and proof-of-principle cohort study. Lancet. 2009;373:1275–1281. doi: 10.1016/S0140-6736(09)60566-3. [DOI] [PubMed] [Google Scholar]
  • 24.Oliveira-Sales EB, Colombari DS, Davisson RL, Kasparov S, Hirata AE, Campos RR, Paton JF. Kidney-induced hypertension depends on superoxide signaling in the rostral ventrolateral medulla. Hypertension. 2010;56:290–296. doi: 10.1161/HYPERTENSIONAHA.110.150425. [DOI] [PubMed] [Google Scholar]
  • 25.Simms AE, Paton JF, Pickering AE, Allen AM. Amplified respiratory-sympathetic coupling in the spontaneously hypertensive rat: does it contribute to hypertension? J Physiol. 2009;587:597–610. doi: 10.1113/jphysiol.2008.165902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sun C, Du J, Sumners C, Raizada MK. PI3-kinase inhibitors abolish the enhanced chronotropic effects of angiotensin II in spontaneously hypertensive rat brain neurons. J Neurophysiol. 2003;90:3155–3160. doi: 10.1152/jn.00222.2003. [DOI] [PubMed] [Google Scholar]
  • 27.Sun C, Zubcevic J, Polson JW, Potts JT, Diez-Freire C, Zhang Q, Paton JF, Raizada MK. Shift to an involvement of phosphatidylinositol 3-kinase in angiotensin II actions on nucleus tractus solitarii neurons of the spontaneously hypertensive rat. Circ Res. 2009;105:1248–1255. doi: 10.1161/CIRCRESAHA.109.208926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Veerasingham SJ, Yamazato M, Berecek KH, Wyss JM, Raizada MK. Increased PI3-kinase in presympathetic brain areas of the spontaneously hypertensive rat. Circ Res. 2005;96:277–279. doi: 10.1161/01.RES.0000156275.06641.b2. [DOI] [PubMed] [Google Scholar]
  • 29.Zubcevic J, Waki H, Diez-Freire C, Gampel A, Raizada MK, Paton JF. Chronic blockade of phosphatidylinositol 3-kinase in the nucleus tractus solitarii is prohypertensive in the spontaneously hypertensive rat. Hypertension. 2009;53:97–103. doi: 10.1161/HYPERTENSIONAHA.108.122341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Simms AE, Paton JF, Allen AM, Pickering AE. Is augmented central respiratory-sympathetic coupling involved in the generation of hypertension? Respir Physiol Neurobiol. 2010;174:89–97. doi: 10.1016/j.resp.2010.07.010. [DOI] [PubMed] [Google Scholar]
  • 31.Zoccal DB, Paton JF, Machado BH. Do changes in the coupling between respiratory and sympathetic activities contribute to neurogenic hypertension? Clin Exp Pharmacol Physiol. 2009;36:1188–1196. doi: 10.1111/j.1440-1681.2009.05202.x. [DOI] [PubMed] [Google Scholar]
  • 32.Zubcevic J, Potts JT. Role of GABAergic neurones in the nucleus tractus solitarii in modulation of cardiovascular activity. Exp Physiol. 2010;95:909–918. doi: 10.1113/expphysiol.2010.054007. [DOI] [PubMed] [Google Scholar]
  • 33.Cachofeiro V, Miana M, de las Heras Natalia, Martin-Fernandez Beatriz, Ballesteros S, Balfagon G, Lahera V. Inflammation: A link between hypertension and atherosclerosis. Curr Hypertens Rev. 2009;5:40–48. [Google Scholar]
  • 34.Sanz-Rosa D, Oubiña MP, Cediel E, de Las Heras N, Vegazo O, Jiménez J, Lahera V, Cachofeiro V. Effect of AT1 receptor antagonism of vascular and circulating inflammatory mediators in SHR: role of NF-kappaB/IkappaB system. Am J Physiol Heart Circ Physiol. 2005;288:H111–H115. doi: 10.1152/ajpheart.01061.2003. [DOI] [PubMed] [Google Scholar]
  • 35.Fliser D, Buchholz K, Haller H. European Trial on Olmesartan and Pravastatin in Inflammation and Atherosclerosis (EUTOPIA) Investigators. Antiinflammatory effects of angiotensin II subtype 1 receptor blockade in hypertensive patients with microinflammation. Circulation. 2004;110:1103–1107. doi: 10.1161/01.CIR.0000140265.21608.8E. [DOI] [PubMed] [Google Scholar]
  • 36.Koh KK, Ahn JY, Han SH, Kim DS, Jin DK, Kim HS, Shin MS, Ahn TH, Choi IS, Shin EK. Pleiotropic effects of angiotensin II receptor blocker in hypertensive patients. J Am Coll Cardiol. 2003;42:905–910. doi: 10.1016/s0735-1097(03)00846-5. [DOI] [PubMed] [Google Scholar]
  • 37.Rahman ST, Lauten WB, Khan QA, Navalkar S, Parthasarathy S, Khan BV. Effects of eprosartan versus hydrochlorothiazide on markers of vascular oxidation and inflammation and blood pressure (renin-angiotensin system antagonists, oxidation, and inflammation). Am J Cardiol. 2002;89:686–690. doi: 10.1016/s0002-9149(01)02340-2. [DOI] [PubMed] [Google Scholar]
  • 38.Schmid-Schönbein GW, Seiffge D, DeLano FA, Shen K, Zweifach BW. Leukocyte counts and activation in spontaneously hypertensive and normotensive rats. Hypertension. 1991;17:323–330. doi: 10.1161/01.hyp.17.3.323. [DOI] [PubMed] [Google Scholar]
  • 39.Fukuda S, Yasu T, Kobayashi N, Ikeda N, Schmid-Schönbein GW. Contribution of fluid shear response in leukocytes to hemodynamic resistance in the spontaneously hypertensive rat. Circ Res. 2004;95:100–108. doi: 10.1161/01.RES.0000133677.77465.38. [DOI] [PubMed] [Google Scholar]
  • 40.Zhang M, Mao Y, Ramirez SH, Tuma RF, Chabrashvili T. Angiotensin II induced cerebral microvascular inflammation and increased blood-brain barrier permeability via oxidative stress. Neuroscience. 2010;171:852–858. doi: 10.1016/j.neuroscience.2010.09.029. [DOI] [PubMed] [Google Scholar]
  • 41.Marvar PJ, Thabet SR, Guzik TJ, Lob HE, McCann LA, Weyand C, Gordon FJ, Harrison DG. Central and peripheral mechanisms of T-lymphocyte activation and vascular inflammation produced by angiotensin II-induced hypertension. Circ Res. 2010;107:263–270. doi: 10.1161/CIRCRESAHA.110.217299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hoch NE, Guzik TJ, Chen W, Deans T, Maalouf SA, Gratze P, Weyand C, Harrison DG. Regulation of T-cell function by endogenously produced angiotensin II. Am J Physiol Regul Integr Comp Physiol. 2009;296:R208–R216. doi: 10.1152/ajpregu.90521.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Boesen EI, Williams DL, Pollock JS, Pollock DM. Immunosuppression with mycophenolate mofetil attenuates the development of hypertension and albuminuria in deoxycorticosterone acetate-salt hypertensive rats. Clin Exp Pharmacol Physiol. 2010;37:1016–1022. doi: 10.1111/j.1440-1681.2010.05428.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ando H, Zhou J, Macova M, Imboden H, Saavedra JM. Angiotensin II AT1 receptor blockade reverses pathological hypertrophy and inflammation in brain microvessels of spontaneously hypertensive rats. Stroke. 2004;35:1726–1731. doi: 10.1161/01.STR.0000129788.26346.18. [DOI] [PubMed] [Google Scholar]
  • 45.Felder RB. Mineralocorticoid receptors, inflammation and sympathetic drive in a rat model of systolic heart failure. Exp Physiol. 2010;95:19–25. doi: 10.1113/expphysiol.2008.045948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Felder RB, Yu Y, Zhang ZH, Wei SG. Pharmacological treatment for heart failure: a view from the brain. Clin Pharmacol Ther. 2009;86:216–220. doi: 10.1038/clpt.2009.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Waki H, Liu B, Miyake M, Katahira K, Murphy D, Kasparov S, Paton JF. Junctional adhesion molecule-1 is upregulated in spontaneously hypertensive rats: evidence for a prohypertensive role within the brain stem. Hypertension. 2007;49:1321–1327. doi: 10.1161/HYPERTENSIONAHA.106.085589. [DOI] [PubMed] [Google Scholar]
  • 48.Waki H, Gouraud SS, Maeda M, Paton JF. Evidence of specific inflammatory condition in nucleus tractus solitarii of spontaneously hypertensive rats. Exp Physiol. 2010;95:595–600. doi: 10.1113/expphysiol.2009.047324. [DOI] [PubMed] [Google Scholar]
  • 49.Kang YM, Ma Y, Zheng JP, Elks C, Sriramula S, Yang ZM, Francis J. Brain nuclear factor-kappa B activation contributes to neurohumoral excitation in angiotensin II-induced hypertension. Cardiovasc Res. 2009;82:503–512. doi: 10.1093/cvr/cvp073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Colombari E, Colombari DS, Li H, Shi P, Dong Y, Jiang N, Raizada MK, Sumners C, Murphy D, Paton JF. Macrophage migration inhibitory factor in the paraventricular nucleus plays a major role in the sympathoexcitatory response to salt. Hypertension. 2010;56:956–963. doi: 10.1161/HYPERTENSIONAHA.110.155101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shi P, Diez-Freire C, Jun JY, Qi Y, Katovich MJ, Li Q, Sriramula S, Francis J, Sumners C, Raizada MK. Brain microglial cytokines in neurogenic hypertension. Hypertension. 2010;56:297–303. doi: 10.1161/HYPERTENSIONAHA.110.150409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kang YM, He RL, Yang LM, Qin DN, Guggilam A, Elks C, Yan N, Guo Z, Francis J. Brain tumour necrosis factor-alpha modulates neurotransmitters in hypothalamic paraventricular nucleus in heart failure. Cardiovasc Res. 2009;83:737–746. doi: 10.1093/cvr/cvp160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kannan H, Tanaka Y, Kunitake T, Ueta Y, Hayashida Y, Yamashita H. Activation of sympathetic outflow by recombinant human interleukin-1 beta in conscious rats. Am J Physiol. 1996;270:R479–R485. doi: 10.1152/ajpregu.1996.270.2.R479. [DOI] [PubMed] [Google Scholar]
  • 54.Lu Y, Chen J, Yin X, Zhao H. Angiotensin II receptor 1 involved in the central pressor response induced by interleukin-1 beta in the paraventricular nucleus. Neurol Res. 2009;31:420–424. doi: 10.1179/174313208X353677. [DOI] [PubMed] [Google Scholar]
  • 55.Yu Y, Zhang ZH, Wei SG, Chu Y, Weiss RM, Heistad DD, Felder RB. Central gene transfer of interleukin-10 reduces hypothalamic inflammation and evidence of heart failure in rats after myocardial infarction. Circ Res. 2007;101:304–312. doi: 10.1161/CIRCRESAHA.107.148940. [DOI] [PubMed] [Google Scholar]
  • 56.Takagishi M, Waki H, Bhuiyan MER, Gouraud SS, Kohsaka A, Cui H, Yamazaki T, Paton JFR, Maeda M. IL-6 microinjected into the nucleus tractus solitarii attenuates cardiac baroreceptor reflex function in rats. Am J Physiol Regul Integr Comp Physiol. 2010;298:R183–R190. doi: 10.1152/ajpregu.00176.2009. [DOI] [PubMed] [Google Scholar]
  • 57.Sun C, Li H, Leng L, Raizada MK, Bucala R, Sumners C. Macrophage migration inhibitory factor: an intracellular inhibitor of angiotensin II-induced increases in neuronal activity. J Neurosci. 2004;24:9944–9952. doi: 10.1523/JNEUROSCI.2856-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kang YM, Ma Y, Elks C, Zheng JP, Yang ZM, Francis J. Cross-talk between cytokines and renin-angiotensin in hypothalamic paraventricular nucleus in heart failure: role of nuclear factor-kappaB. Cardiovasc Res. 2008;79:671–678. doi: 10.1093/cvr/cvn119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Peterson JR, Sharma RV, Davisson RL. Reactive oxygen species in the neuropathogenesis of hypertension. Curr Hypertens Rep. 2006;8:232–241. doi: 10.1007/s11906-006-0056-1. [DOI] [PubMed] [Google Scholar]
  • 60.Li DP, Chen SR, Finnegan TF, Pan HL. Signalling pathway of nitric oxide in synaptic GABA release in the rat paraventricular nucleus. J Physiol. 2004;554:100–110. doi: 10.1113/jphysiol.2003.053371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Utsuyama M, Hirokawa K. Differential expression of various cytokine receptors in the brain after stimulation with LPS in young and old mice. Exp Gerontol. 2002;37:411–420. doi: 10.1016/s0531-5565(01)00208-x. [DOI] [PubMed] [Google Scholar]
  • 62.Dickinson CJ, Thomson AD. A post mortem study of the main cerebral arteries with special reference to their possible role in blood pressure regulation. Clin Sci. 1960;19:513–538. [PubMed] [Google Scholar]
  • 63.Paton JF, Waki H, Abdala AP, Dickinson J, Kasparov S. Vascular-brain signaling in hypertension: role of angiotensin II and nitric oxide. Curr Hypertens Rep. 2007;9:242–247. doi: 10.1007/s11906-007-0043-1. [DOI] [PubMed] [Google Scholar]
  • 64.Yenari MA, Xu L, Tang XN, Qiao Y, Giffard RG. Microglia potentiate damage to blood-brain barrier constituents: improvement by minocycline in vivo and in vitro. Stroke. 2006;37:1087–1093. doi: 10.1161/01.STR.0000206281.77178.ac. [DOI] [PubMed] [Google Scholar]
  • 65.Saavedra JM, Sánchez-Lemus E, Benicky J. Blockade of brain angiotensin II AT1 receptors ameliorates stress, anxiety, brain inflammation and ischemia: Therapeutic implications. Psychoneuroendocrinology. 2011;36:1–18. doi: 10.1016/j.psyneuen.2010.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Perry VH, Nicoll JA, Holmes C. Microglia in neurodegenerative disease. Nat Rev Neurol. 2010;6:193–201. doi: 10.1038/nrneurol.2010.17. [DOI] [PubMed] [Google Scholar]
  • 67.Appel SH, Beers DR, Henkel JS. T cell-microglial dialogue in Parkinson's disease and amyotrophic lateral sclerosis: are we listening? Trends Immunol. 2010;31:7–17. doi: 10.1016/j.it.2009.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Benicky J, Sánchez-Lemus E, Honda M, Pang T, Orecna M, Wang J, Leng Y, Chuang DM, Saavedra JM. Angiotensin II AT(1) Receptor Blockade Ameliorates Brain Inflammation. Neuropsychopharmacology. 2011;36:857–870. doi: 10.1038/npp.2010.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Benicky J, Sánchez-Lemus E, Pavel J, Saavedra JM. Anti-inflammatory effects of angiotensin receptor blockers in the brain and the periphery. Cell Mol Neurobiol. 2009;29:781–792. doi: 10.1007/s10571-009-9368-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Carnevale D, Mascio G, Ajmone-Cat MA, D'Andrea I, Cifelli G, Madonna M, Cocozza G, Frati A, Carullo P, Carnevale L, Alleva E, Branchi I, Lembo G, Minghetti L. Role of neuroinflammation in hypertension-induced brain amyloid pathology. Neurobiol Aging. 2010 doi: 10.1016/j.neurobiolaging.2010.08.013. Epub ahead of print. PMID: 20961666. [DOI] [PubMed] [Google Scholar]
  • 71.Lanz TV, Ding Z, Ho PP, Luo J, Agrawal AN, Srinagesh H, Axtell R, Zhang H, Platten M, Wyss-Coray T, Steinman L. Angiotensin II sustains brain inflammation in mice via TGF-beta. J Clin Invest. 2010;120:2782–2794. doi: 10.1172/JCI41709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Borovikova LV, Ivanova S, Nardi D, Zhang M, Yang H, Ombrellino M, Tracey KJ. Role of vagus nerve signaling in CNI-1493-mediated suppression of acute inflammation. Auton Neurosci. 2000;85:141–147. doi: 10.1016/S1566-0702(00)00233-2. [DOI] [PubMed] [Google Scholar]
  • 73.Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH, Wang H, Yang H, Ulloa L, Al-Abed Y, Czura CJ, Tracey KJ. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 2003;421:384–388. doi: 10.1038/nature01339. [DOI] [PubMed] [Google Scholar]
  • 74.de Jonge WJ, van der Zanden EP, The FO, Bijlsma MF, van Westerloo DJ, Bennink RJ, Berthoud HR, Uematsu S, Akira S, van den Wijngaard RM, Boeckxstaens GE. Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat Immunol. 2005;6:844–851. doi: 10.1038/ni1229. [DOI] [PubMed] [Google Scholar]
  • 75.Ek M, Kurosawa M, Lundeberg T, Ericsson A. Activation of vagal afferents after intravenous injection of interleukin-1beta: role of endogenous prostaglandins. J Neurosci. 1998;18:9471–9479. doi: 10.1523/JNEUROSCI.18-22-09471.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Goehler LE, Relton JK, Dripps D, Kiechle R, Tartaglia N, Maier SF, Watkins LR. Vagal paraganglia bind biotinylated interleukin-1 receptor antagonist: a possible mechanism for immune-to-brain communication. Brain Res Bull. 1997;43:357–364. doi: 10.1016/s0361-9230(97)00020-8. [DOI] [PubMed] [Google Scholar]
  • 77.Goehler LE, Gaykema RP, Hansen MK, Anderson K, Maier SF, Watkins LR. Vagal immune-to-brain communication: a visceral chemosensory pathway. Auton Neurosci. 2000;85:49–59. doi: 10.1016/S1566-0702(00)00219-8. [DOI] [PubMed] [Google Scholar]
  • 78.Tracey KJ. The inflammatory reflex. Nature. 2002;420:853–859. doi: 10.1038/nature01321. [DOI] [PubMed] [Google Scholar]
  • 79.Tracey KJ. Understanding immunity requires more than immunology. Nature Immunol. 2010;11:561–564. doi: 10.1038/ni0710-561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.La Rovere MT, Mortara A, Pantaleo P, Maestri R, Cobelli F, Tavazzi L. Scopolamine improves autonomic balance in advanced congestive heart failure. Circulation. 1994;90:838–843. doi: 10.1161/01.cir.90.2.838. [DOI] [PubMed] [Google Scholar]
  • 81.Halliwill JR, Billman GE, Eckberg DL. Effect of a ‘vagomimetic’ atropine dose on canine cardiac vagal tone and susceptibility to sudden cardiac death. Clin Auton Res. 1998;8:155–164. doi: 10.1007/BF02281120. [DOI] [PubMed] [Google Scholar]
  • 82.Yao EH, Yu Y, Fukuda N. Oxidative stress on progenitor and stem cells in cardiovascular diseases. Curr Pharm Biotechnol. 2006;7:101–108. doi: 10.2174/138920106776597685. [DOI] [PubMed] [Google Scholar]
  • 83.Giannotti G, Doerries C, Mocharla PS, Mueller MF, Bahlmann FH, Horvàth T, Jiang H, Sorrentino SA, Steenken N, Manes C, Marzilli M, Rudolph KL, Lüscher TF, Drexler H, Landmesser U. Impaired endothelial repair capacity of early endothelial progenitor cells in prehypertension: relation to endothelial dysfunction. Hypertension. 2010;55:1389–1397. doi: 10.1161/HYPERTENSIONAHA.109.141614. [DOI] [PubMed] [Google Scholar]
  • 84.MacEneaney OJ, DeSouza CA, Weil BR, Kushner EJ, Van Guilder GP, Mestek ML, Greiner JJ, Stauffer BL. Prehypertension and endothelial progenitor cell function. J Hum Hypertens. 2011;25:57–62. doi: 10.1038/jhh.2010.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Imanishi T, Hano T, Nishio I. Angiotensin II accelerates endothelial progenitor cell senescence through induction of oxidative stress. J Hypertens. 2005;23:97–104. doi: 10.1097/00004872-200501000-00018. [DOI] [PubMed] [Google Scholar]
  • 86.Imanishi T, Moriwaki C, Hano T, Nishio I. Endothelial progenitor cell senescence is accelerated in both experimental hypertensive rats and patients with essential hypertension. J Hypertens. 2005;23:1831–1837. doi: 10.1097/01.hjh.0000183524.73746.1b. [DOI] [PubMed] [Google Scholar]
  • 87.Jialal I, Fadini GP, Pollock K, Devaraj S. Circulating levels of endothelial progenitor cell mobilizing factors in the metabolic syndrome. Am J Cardiol. 2010;106:1606–1608. doi: 10.1016/j.amjcard.2010.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Jarajapu YP, Grant MB. The promise of cell-based therapies for diabetic complications: challenges and solutions. Circ Res. 2010;106:854–869. doi: 10.1161/CIRCRESAHA.109.213140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tongers J, Losordo DW. Frontiers in nephrology: the evolving therapeutic applications of endothelial progenitor cells. J Am Soc Nephrol. 2007;18:2843–2852. doi: 10.1681/ASN.2007050597. [DOI] [PubMed] [Google Scholar]
  • 90.van Zonneveld AJ, de Boer HC, van der Veer EP, Rabelink TJ. Inflammation, vascular injury and repair in rheumatoid arthritis. Ann Rheum Dis. 2010;69(Suppl 1):i57–i60. doi: 10.1136/ard.2009.119495. [DOI] [PubMed] [Google Scholar]
  • 91.Huang PH, Huang SS, Chen YH, Lin CP, Chiang KH, Chen JS, Tsai HY, Lin FY, Chen JW, Lin SJ. Increased circulating CD31+/annexin V+ apoptotic microparticles and decreased circulating endothelial progenitor cell levels in hypertensive patients with microalbuminuria. J Hypertens. 2010;28:1655–1665. doi: 10.1097/HJH.0b013e32833a4d0a. [DOI] [PubMed] [Google Scholar]
  • 92.Müller P, Kazakov A, Jagoda P, Semenov A, Böhm M, Laufs U. ACE inhibition promotes upregulation of endothelial progenitor cells and neoangiogenesis in cardiac pressure overload. Cardiovasc Res. 2009;83:106–114. doi: 10.1093/cvr/cvp123. [DOI] [PubMed] [Google Scholar]
  • 93.Honda A, Matsuura K, Fukushima N, Tsurumi Y, Kasanuki H, Hagiwara N. Telmisartan induces proliferation of human endothelial progenitor cells via PPARgamma-dependent PI3K/Akt pathway. Atherosclerosis. 2009;205:376–384. doi: 10.1016/j.atherosclerosis.2008.12.036. [DOI] [PubMed] [Google Scholar]
  • 94.Serre CM, Farlay D, Delmas PD, Chenu C. Evidence for a dense and intimate innervation of the bone tissue, including glutamate-containing fibers. Bone. 1999;25:623–629. doi: 10.1016/s8756-3282(99)00215-x. [DOI] [PubMed] [Google Scholar]
  • 95.Mignini F, Streccioni V, Amenta F. Autonomic innervation of immune organs and neuroimmune modulation. Auton Autocoid Pharmacol. 2003;23:1–25. doi: 10.1046/j.1474-8673.2003.00280.x. [DOI] [PubMed] [Google Scholar]
  • 96.Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, Frenette PS. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407–421. doi: 10.1016/j.cell.2005.10.041. [DOI] [PubMed] [Google Scholar]
  • 97.Busik JV, Tikhonenko M, Bhatwadekar A, Opreanu M, Yakubova N, Caballero S, Player D, Nakagawa T, Afzal A, Kielczewski J, Sochacki A, Hasty S, Li Calzi S, Kim S, Duclas SK, Segal MS, Guberski DL, Esselman WJ, Boulton ME, Grant MB. Diabetic retinopathy is associated with bone marrow neuropathy and a depressed peripheral clock. J Exp Med. 2009;206:2897–2906. doi: 10.1084/jem.20090889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Artico M, Bosco S, Cavallotti C, Agostinelli E, Giuliani-Piccari G, Sciorio S, Cocco L, Vitale M. Noradrenergic and cholinergic innervation of the bone marrow. Int J Mol Med. 2002;10:77–80. [PubMed] [Google Scholar]
  • 99.Dénes A, Boldogkoi Z, Uhereczky G, Hornyák A, Rusvai M, Palkovits M, Kovács KJ. Central autonomic control of the bone marrow: multisynaptic tract tracing by recombinant pseudorabies virus. Neuroscience. 2005;134:947–963. doi: 10.1016/j.neuroscience.2005.03.060. [DOI] [PubMed] [Google Scholar]
  • 100.Yoshimoto M, Miki K, Fink GD, King A, Osborn JW. Chronic angiotensin II infusion causes differential responses in regional sympathetic nerve activity in rats. Hypertension. 2010;55:644–651. doi: 10.1161/HYPERTENSIONAHA.109.145110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Osborn JW, Fink GD. Region-specific changes in sympathetic nerve activity in angiotensin II-salt hypertension in the rat. Exp Physiol. 2010;95:61–68. doi: 10.1113/expphysiol.2008.046326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ferguson AV. Angiotensinergic regulation of autonomic and neuroendocrine outputs: critical roles for the subfornical organ and paraventricular nucleus. Neuroendocrinology. 2009;89:370–376. doi: 10.1159/000211202. [DOI] [PubMed] [Google Scholar]
  • 103.Davisson RL, Zimmerman MC. Angiotensin II, oxidant signaling, and hypertension: down to a T? Hypertension. 2010;55:228–230. doi: 10.1161/HYPERTENSIONAHA.109.144477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Pelisch N, Hosomi N, Ueno M, Nakano D, Hitomi H, Mogi M, Shimada K, Kobori H, Horiuchi M, Sakamoto H, Matsumoto M, Kohno M, Nishiyama A. Blockade of AT1 receptors protects the blood-brain barrier and improves cognition in Dahl salt-sensitive hypertensive rats. Am J Hypertens. 2011;24:362–368. doi: 10.1038/ajh.2010.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Vital SA, Terao S, Nagai M, Granger DN. Mechanisms underlying the cerebral microvascular responses to angiotensin II-induced hypertension. Microcirculation. 2010;17:641–649. doi: 10.1111/j.1549-8719.2010.00060.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ueno M, Sakamoto H, Liao YJ, Onodera M, Huang CL, Miyanaka H, Nakagawa T. Blood-brain barrier disruption in the hypothalamus of young adult spontaneously hypertensive rats. Histochem Cell Biol. 2004;122:131–137. doi: 10.1007/s00418-004-0684-y. [DOI] [PubMed] [Google Scholar]
  • 107.Poulet R, Gentile MT, Vecchione C, Distaso M, Aretini A, Fratta L, Russo G, Echart C, Maffei A, De Simoni MG, Lembo G. Acute hypertension induces oxidative stress in brain tissues. J Cereb Blood Flow Metab. 2006;26:253–262. doi: 10.1038/sj.jcbfm.9600188. [DOI] [PubMed] [Google Scholar]
  • 108.Chen SK, Tvrdik P, Peden E, Cho S, Wu S, Spangrude G, Capecchi MR. Hematopoietic origin of pathological grooming in Hoxb8 mutant mice. Cell. 2010;141:775–785. doi: 10.1016/j.cell.2010.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Longo B, Romariz S, Blanco MM, Vasconcelos JF, Bahia L, Soares MB, Mello LE, Ribeiro-dos-Santos R. Distribution and proliferation of bone marrow cells in the brain after pilocarpine-induced status epilepticus in mice. Epilepsia. 2010;51:1628–1632. doi: 10.1111/j.1528-1167.2010.02570.x. [DOI] [PubMed] [Google Scholar]
  • 110.Cato MJ, Toney GM. Angiotensin II excites paraventricular nucleus neurons that innervate the rostral ventrolateral medulla: an in vitro patch-clamp study in brain slices. J Neurophysiol. 2005;93:403–413. doi: 10.1152/jn.01055.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pfister J, Spengler C, Grouzmann E, Raizada MK, Felix D, Imboden H. Intracellular staining of angiotensin receptors in the PVN and SON of the rat. Brain Res. 1997;754:307–310. doi: 10.1016/s0006-8993(97)00180-7. [DOI] [PubMed] [Google Scholar]

RESOURCES