Elsevier

Nitric Oxide

Volume 94, 1 January 2020, Pages 54-62
Nitric Oxide

Central angiotensin II-Protein inhibitor of neuronal nitric oxide synthase (PIN) axis contribute to neurogenic hypertension

https://doi.org/10.1016/j.niox.2019.10.007Get rights and content

Highlights

  • Intracerebroventricular (icv) infusion of Ang II increases the expression of PIN in the PVN.

  • Ubiquitination of PIN is decreased in NG108 cells treated with Ang II and also in the PVN of rats infused with AngII (icv).

  • Ang II via the Ubiquitin-Proteasomal pathway may regulate PIN expression in the PVN at the post-translational level.

  • The increased PIN would be expected to decrease active nNOS dimers and hence reduce the production of nitric oxide.

  • The reduced NO• levels would lead to abrogated inhibition of sympathetic tone mediated from the PVN.

Abstract

Activation of renin-angiotensin- system, nitric oxide (NO•) bioavailability and subsequent sympathoexcitation plays a pivotal role in the pathogenesis of many cardiovascular diseases, including hypertension. Previously we have shown increased protein expression of PIN (a protein inhibitor of nNOS: neuronal nitric oxide synthase, known to dissociate nNOS dimers into monomers) with concomitantly reduced levels of catalytically active dimers of nNOS in the PVN of rats with heart failure. To elucidate the molecular mechanism by which Angiotensin II (Ang II) increases PIN expression, we used Sprague-Dawley rats (250–300 g) subjected to intracerebroventricular infusion of Ang II (20 ng/min, 0.5 μl/h) or saline as vehicle (Veh) for 14 days through osmotic mini-pumps and NG108-15 hybrid neuronal cell line treated with Ang II as an in vitro model. Ang II infusion significantly increased baseline renal sympathetic nerve activity and mean arterial pressure. Ang II infusion increased the expression of PIN (1.24 ± 0.04* Ang II vs. 0.65 ± 0.07 Veh) with a concomitant 50% decrease in dimeric nNOS and PIN-Ub conjugates (0.73 ± 0.04* Ang II vs. 1.00 ± 0.03 Veh) in the PVN. Substrate-dependent ligase assay in cells transfected with pCMV-(HA-Ub)8 vector revealed a reduction of HA-Ub-PIN conjugates after Ang II and a proteasome inhibitor, Lactacystin (LC), treatment (4.5 ± 0.7* LC Ang II vs. 9.2 ± 2.5 LC). TUBE (Tandem Ubiquitin-Binding Entities) assay showed decrease PIN-Ub conjugates in Ang II-treated cells (0.82 ± 0.12* LC Ang II vs. 1.21 ± 0.06 LC) while AT1R blocker, Losartan (Los) treatment diminished the Ang II-mediated stabilization of PIN (1.21 ± 0.07 LC Los vs. 1.16 ± 0.04* LC Ang II Los). Taken together, our studies suggest that increased central levels of Ang II contribute to the enhanced expression of PIN leading to reduced expression of the dimeric form of nNOS, thus diminishing the inhibitory action of NO• on pre-autonomic neurons in the PVN resulting in increased sympathetic outflow.

Introduction

Reduced bioavailability of nitric oxide (NO•) concomitant with increased sympathetic outflow is implicated in the pathogenesis of many cardiovascular diseases, including hypertension, heart failure, and diabetes [[1], [2], [3], [4], [5]]. Further, hypertensive patients and animal models of hypertension have demonstrated increased sympathetic nerve activity (SNA), accompanied by alterations in body fluid homeostasis [6]. The NO•-pathway contributes to the blood pressure-lowering effect of many commonly used therapeutic agents and emerging basic science research on this pathway is elucidating potential new antihypertensive drug targets. NO• is a gaseous paracrine molecule produced by a family of NADPH-dependent enzymes named nitric oxide synthases (NOS) [[7], [8], [9]]. NO• acts as a pivotal signaling molecule that regulates blood flow and tissue oxygenation by activating soluble guanylate cyclase in the vascular smooth muscle [10]. NO• produced by the neuronal isoform of NOS (nNOS) plays an essential role in the paraventricular nucleus (PVN) via acting as a retrograde inhibitory neurotransmitter that regulates synaptic efficacy and modulates neuronal activity leading to changes in SNA and consequently neurogenic control of blood pressure [[11], [12], [13]]. Of the three NOS isoforms, nNOS is the largest, due to a 300-amino-acid PDZ (PSD-95/Discs large/zona occludens-1) domain insertion at the N-terminus targeting nNOS to the postsynaptic density (PSD) protein in the synaptic spines of neurons [14]. The catalytic activity of nNOS is regulated by several other scaffolding proteins such as syntrophin, PSD93, PSD95 and CAPON (carboxy-terminal PDZ ligand of nNOS) which have been identified to interact with nNOS in neurons and modify its activity. In neurons, another nNOS-interacting protein the dynein light chain, LC8 (DYNLL1) or PIN (protein inhibitor of nNOS) physically interacts with nNOS and inhibits its activity [15]. PIN is a highly conserved, small homodimer cytosolic protein that is reported to bind to a 17-residue peptide fragment from Met-228 to His-244 of nNOS [16] and destabilizes the nNOS homodimer [15], a conformation necessary for the catalytic activity of the enzyme. Monomerization of nNOS promotes ubiquitination and degradation of nNOS by the proteasomal pathway [17,18]. Previously, we have shown that there is a decrease in nNOS expression [19] and enhanced expression of CAPON [20] and PIN [4] in the PVN of rats with heart failure. Moreover, Losartan (Los) an AT1R blocker treatment restores the elevated SNA and normalizes the changes in expression of nNOS, CAPON, and PIN suggesting that the AT1 receptor activation may be linked to the reciprocal changes in these regulators(CAPON and PIN) and nNOS in the PVN [4,20]. Furthermore, we described the in vitro molecular mechanism of downregulation of nNOS involving interactions between nNOS and PIN through an Ang II-mediated signaling pathway(s) and suggested that post-translational processes, such as protein degradation/stabilization are involved in Ang II-dependent downregulation of nNOS [4]. To further understand how Ang II to PIN link, regulates nNOS, we dissect the molecular mechanism of increased PIN expression in a well-established model of neurogenic hypertension produced by intracerebroventricular infusion of Ang II for two weeks.

Section snippets

Animal model

Male Sprague-Dawley rats weighing between 250 and 280 g (Sasco Breeding Laboratories, Omaha, NE) were fed and housed according to institutional guidelines. The University of Nebraska Medical Center Institutional Animal Care and Use Committee approved all protocols (IACUC protocol # 14-036-07-FC) which adhere to the National Institutes of Health Guide for the Care and Use of Laboratory Animals, Eighth Edition (National Academic Press; 2011).

Intracerebroventricular (ICV) infusion of Ang II

Rats were randomly assigned to one of two groups:

Results

In the present study, we used the ICV infusion of Ang II as a model of hypertension, which has been previously used by numerous investigators to produce hypertension with evidence for a strong contribution of the neurogenic component [[28], [29], [30], [31]]. As anticipated, ICV infusion of Ang II-induced an increase in mean arterial pressure compared to vehicle group (126 ± 9 Ang II vs. 84 ± 4 mmHg) and increased baseline RSNA (20.5 ± 2.3 Ang II vs. 6.4 ± 1.9% n = 5 and 4, respectively of

Discussion

The salient findings of this study are that central infusion of Ang II; 1) increases RSNA and MAP, 2) increases PIN expression, and decreases the ubiquitination of PIN within the PVN, and 3) decreases the dimeric level of nNOS and hence decrease NO• bioavailability. Further, data showing no effects of Ang II on mRNA expression and new protein synthesis of PIN indicate that protein degradation may be the primary mechanism for the upregulation of PIN by Ang II. These results suggest that central

Declaration of competing interest

The authors declare no potential conflicts of interest.

Acknowledgments

Funding from American Heart Association National Center grant 14SDG19980007(N. Sharma) and National Institutes of Health, Heart, Lung, & Blood Institute, Grant R01-DK-114663, HL62222 and McIntyre Professorship (K.P. Patel) supported this work.

References (70)

  • S.C. Wang et al.

    RNA silencing targeting PIN (protein inhibitor of neuronal nitric oxide synthase) attenuates the development of hypertension in young spontaneously hypertensive rats

    J Am Soc Hypertens

    (2014)
  • K.J. Catt et al.

    Angiotensin II blood-levels in human hypertension

    Lancet

    (1971)
  • P.C. Perin et al.

    Sympathetic nervous system, diabetes, and hypertension

    Clin. Exp. Hypertens.

    (2001)
  • I.H. Zucker et al.

    The regulation of sympathetic outflow in heart failure. The roles of angiotensin II, nitric oxide, and exercise training

    Ann. N. Y. Acad. Sci.

    (2001)
  • H. Zheng et al.

    Exercise training normalizes enhanced sympathetic activation from the paraventricular nucleus in chronic heart failure: role of angiotensin II

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2012)
  • N.M. Sharma et al.

    Angiotensin II-mediated posttranslational modification of nNOS in the PVN of rats with CHF: role for PIN

    Am. J. Physiol. Heart Circ. Physiol.

    (2013)
  • F. Fyhrquist et al.

    Role of angiotensin II in blood pressure regulation and in the pathophysiology of cardiovascular disorders

    J. Hum. Hypertens.

    (1995)
  • P. Palatini et al.

    The role of cardiac autonomic function in hypertension and cardiovascular disease

    Curr. Hypertens. Rep.

    (2009)
  • A.C. Gorren et al.

    The versatile and complex enzymology of nitric oxide synthase

    Biochemistry (Mosc.)

    (1998)
  • E.R. Werner et al.

    Tetrahydrobiopterin, cytokines, and nitric oxide synthesis

    Proc. Soc. Exp. Biol. Med.

    (1998)
  • K. Chen et al.

    Nitric oxide in the vasculature: where does it come from and where does it go? A quantitative perspective

    Antioxidants Redox Signal.

    (2008)
  • K. Zhang et al.

    Nitric oxide within the paraventricular nucleus mediates changes in renal sympathetic nerve activity

    Am. J. Physiol.

    (1997)
  • K. Zhang et al.

    Effect of nitric oxide within the paraventricular nucleus on renal sympathetic nerve discharge:role of GABA

    Am. J. Physiol.

    (1998)
  • F.D. McBryde et al.

    Hypothalamic paraventricular nucleus neuronal nitric oxide synthase activity is a major determinant of renal sympathetic discharge in conscious Wistar rats

    Exp. Physiol.

    (2018)
  • S.R. Jaffrey et al.

    PIN: an associated protein inhibitor of neuronal nitric oxide synthase

    Science

    (1996)
  • A.Y. Dunbar et al.

    Ubiquitination and degradation of neuronal nitric-oxide synthase in vitro: dimer stabilization protects the enzyme from proteolysis

    Mol. Pharmacol.

    (2004)
  • Y.-F. Li et al.

    Paraventricular nucleus of the hypothalamus and elevated sympathetic activity in heart failure: altered inhibtory mechanisms

    Acta Physiol. Scand.

    (2003)
  • N.M. Sharma et al.

    Decreased nNOS in the PVN leads to increased sympathoexcitation in chronic heart failure: role for CAPON and Ang II

    Cardiovasc. Res.

    (2011)
  • G. Paxinos et al.

    The Rat Brain in Stereotaxic Coordinates

    (1986)
  • Y.F. Li et al.

    Angiotensin-mediated increase in renal sympathetic nerve discharge within the PVN: role of nitric oxide

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2006)
  • M. Palkovits et al.

    Brain microdissection techniques

  • P. Klatt et al.

    Structural analysis of porcine brain nitric oxide synthase reveals a role for tetrahydrobiopterin and L-arginine in the formation of an SDS-resistant dimer

    EMBO J.

    (1995)
  • R. Hjerpe et al.

    Efficient protection and isolation of ubiquitylated proteins using tandem ubiquitin-binding entities

    EMBO Rep.

    (2009)
  • M. Furukawa et al.

    Ubiquitin proteosome protocols

    Methods Mol. Biol.

    (2005)
  • T.E. Lohmeier

    Angiotensin II infusion model of hypertension: is there an important sympathetic component?

    Hypertension

    (2012)
  • Cited by (10)

    • The heart is lost without the hypothalamus

      2021, Handbook of Clinical Neurology
      Citation Excerpt :

      Studies have revealed the posttranslational mechanism of increased ubiquitination-mediated degradation of NOS1 in the PVN of HF rats leads to sympathoexcitation and ANGII alters the activity of one of three enzymes necessary to ubiquitinate the PIN protein. The action of ANGII on intracellular PIN-ubiquitination means PIN remains within the cell for a longer period of time and transcription of PIN remains unchanged (Sharma et al., 2020). The interesting point here is that PIN is normally targeted for rapid ubiquitin degradation and by this mechanism NOS1 levels and thus NO availability within neurons remain stable.

    • Nanoformulation of the superoxide dismutase mimic, MnTnBuOE-2-PyP<sup>5+</sup>, prevents its acute hypotensive response

      2020, Redox Biology
      Citation Excerpt :

      Rats were housed in the animal facility with a 12 h light-dark cycle with access to standard chow and water ad libitium. Upon arrival rats were allowed to acclimate for one week before RSNA was recorded, as previously described [26,27]. Briefly, rats were anesthetized with an IP injection of urethane (0.75 g/kg) and α-chloralose (70 mg/kg).

    View all citing articles on Scopus
    View full text