Elsevier

Behavioural Brain Research

Volume 154, Issue 1, 23 September 2004, Pages 239-243
Behavioural Brain Research

Research report
Attenuation of scopolamine-induced learning deficits by LVV-hemorphin-7 in rats in the passive avoidance and water maze paradigms

https://doi.org/10.1016/j.bbr.2004.02.012Get rights and content

Abstract

Central administration of angiotensin IV (Ang IV) analogues attenuates scopolamine-induced amnesia. Ang IV mediates its effects by binding to a high affinity, binding site, AT4 receptor, that has recently been identified as insulin regulated aminopeptidase (IRAP). The purpose of this study was to examine the effect of the distinct AT4 ligand, LVV-hemorphin-7 (LVV-H7), on scopolamine-induced learning deficits, one which involves fear-conditioning and the other spatial learning. Rats were pretreated with an intracerebroventricular (ICV) dose of scopolamine hydrobromide followed by treatment with 1 nmol LVV-H7 or artificial cerebrospinal fluid (aCSF). During the acquisition phase of the water maze task, daily ICV infusions of 1 nmol of LVV-H7 25 min after scopolamine treatment produced marked improvement in both the latency and distance swum in order to locate the submerged platform using visual cues compared to animals treated with scopolamine only. In addition, the same dose of LVV-H7 attenuated the learning deficit observed for scopolamine-treated animals in the passive avoidance task. These studies clearly demonstrate that LVV-H7, like Ang IV, is a pharmacologically active AT4 ligand that attenuates the deleterious effects of scopolamine on learning performance in two different behavioral paradigms.

Introduction

Angiotensin IV (Ang IV), a hexapeptide corresponding to residues 3–8 of angiotensin II (Ang II), mediates biological effects that are independent of the classical angiotensin system [29]. Ang IV binds with high affinity to a pharmacologically distinct binding site, termed the AT4 receptor [5], [6]. We have recently identified the AT4 receptor as the transmembrane enzyme insulin regulated aminopeptidase (IRAP) via mass spectral analysis of tryptic peptides generated from AT4 receptor protein purified from bovine adrenal membranes [1]. Analysis of the biochemical and pharmacological properties of IRAP confirm that it is the AT4 receptor.

Both acute and chronic infusions of Ang IV or its stable analogues, Nle1-Ang IV and Nle1-γ-Tyr2-Ang IV (where γCH2NH bond) into the cerebral ventricles has been shown to facilitate memory retention and retrieval in rats in both the passive avoidance and water maze paradigms [31]. In two rat models of amnesia, induced by either central scopolamine administration or bilateral perforant pathway lesion, the Ang IV analogues reversed performance deficits detected in the water maze paradigm [22], [23], [31]. These effects are thought to be mediated by binding of the Ang IV peptides to the AT4 receptor or IRAP.

The AT4 receptor/IRAP has a distinct distribution pattern in the brain and is found in regions associated with cognitive, sensory and motor function. In the human brain, high levels of IRAP are found in the basal nucleus of Meynert, in the CA1 to CA3 regions of the hippocampus, and throughout the neocortex, a distribution that closely resembles cholinergic neurons and their projections [4]. In comparative studies we observe that the distribution of IRAP is highly conserved in the brains of sheep [18], macaque monkey [20] and guinea pig [17] and is abundant in many of the same regions as in the human brain. This is consistent with the demonstrated role of AT4 receptor/IRAP in facilitating memory and learning.

We have previously isolated a decapeptide, LVV-hemorphin-7 (LVV-H7), from the sheep cerebral cortex [19] which bound with high affinity to IRAP. This AT4 ligand, although structurally distinct from Ang IV, exhibits many of the same effects as the hexapeptide in a number of in vitro assays including stimulating cellular proliferation [21], facilitating K+-evoked acetylcholine release in hippocampal slices [14] and competitively inhibiting the catalytic activity of IRAP [1]. However, the ability of LVV-H7 to overcome cognitive deficits in vivo has not been explored. In the present study we investigated the effect of LVV-H7 on memory deficits induced by scopolamine utilizing two behavioural paradigms, the passive avoidance conditioning and the water maze tasks.

Section snippets

Animals and surgery

Male Sprague–Dawley rats (230–280 g), obtained from Animal Resource Centre, Western Australia, were housed individually in an animal housing facility maintained at 20 °C, with an alternating 12:12 h light/dark cycle initiated at 07:00 h. All animals had ad libitum access to water and standard rat chow. Each animal was anaesthetized with Equithesin (3.0 ml/kg administered 1:1 with normal saline) and an intracerebroventricular (ICV) cannula (23-gauge needle) was stereotaxically implanted above the

Water maze

A statistically significant treatment effect was observed in both latency (F2,140=27.271, P<0.001) and swim distance (F2,148=28.839, P<0.001) to find the submerged platform. Post hoc analysis revealed that during the days of acquisition (days 3–7), the 70 nmol scopolamine-treated rats took significantly longer to find the submerged platform (t=6.698, P<0.001) and travelled longer distances (t=4.391, P<0.001) compared to members of the control group. Rats treated with 1 nmol LVV-H7 after

Discussion

This study clearly demonstrates that LVV-H7 attenuates the deleterious effects of scopolamine on learning performance in two different behavioral paradigms, which involves fear-conditioning and place learning. During the acquisition phase of the water maze task, daily intracerebroventricular infusions of 1 nmol of LVV-H7 25 min after scopolamine treatment produced marked improvement in both the latency and distance swum in order to locate the submerged platform using visual cues. This improvement

Acknowledgements

This work is supported by an NHMRC block grant (#983001).

References (31)

Cited by (62)

  • Cognitive benefits of angiotensin IV and angiotensin-(1–7): A systematic review of experimental studies

    2018, Neuroscience and Biobehavioral Reviews
    Citation Excerpt :

    This AT4R ligand is hypothesized to exert its effects through modulation of the central cholinergic system, given the correlation of IRAP with cholinergic cell bodies and terminals (Chai et al., 2000) and the insulin-regulated glucose transporter (GLUT4) in the brain. It has been suggested that LVV-H7 may potentiate hippocampal ACh release through promoting the uptake of glucose into neurons (Albiston et al., 2004). Intrahippocampal injections of glucose increase hippocampal ACh release in rats, an effect linked to enhanced cognitive performance (Ragozzino et al., 1996; Ragozzino et al., 1998).

  • Forebrain neurone-specific deletion of insulin-regulated aminopeptidase causes age related deficits in memory

    2016, Neurobiology of Learning and Memory
    Citation Excerpt :

    Insulin-regulated aminopeptidase (IRAP) is the AT4 receptor, the binding site for the AT4 ligands angiotensin IV (Ang IV, VYIHPF) and LVV-hemorphin 7 (LVV-H7, LVVYPWTQRF) (Albiston et al., 2001). Central infusion of AT4 ligands improves memory in both normal rodents (Lee et al., 2004; Wright et al., 1999) and in models of memory deficit (Albiston et al., 2004; Wright et al., 1999). Other binding sites have been proposed to mediate the cognitive effects of Ang IV and its analogues (Wright & Harding, 2011), but we recently demonstrated the complete absence of Ang IV binding in IRAP knockout mouse tissues (Albiston et al., 2010), confirming IRAP as the primary site of action.

  • The development of small molecule angiotensin IV analogs to treat Alzheimer's and Parkinson's diseases

    2015, Progress in Neurobiology
    Citation Excerpt :

    In fact, central blockade of the AT1 receptor by ARBs has been shown to facilitate a variety of memory tasks in rats and mice (Basso et al., 2005; Bild et al., 2013; Ellul et al., 2006; Hajjar et al., 2005; Khachaturian et al., 2006; Marvar et al., 2013; Mogi and Horiuchi, 2009); while central administration of AngII impaired spatial memory (Tota et al., 2013). A number of studies have noted AngIV and AngIV analog facilitation of LTP and memory consolidation (Albiston et al., 2004; Braszko et al., 1988; Kramár et al., 2001; Lee et al., 2004; Paris et al., 2013; Wayner et al., 2001; Wright et al., 1999). Central administration of AngIV (Braszko et al., 1988, 2008; De Bundel et al., 2009; Paris et al., 2013; Wright et al., 1993, 1996a), or the AngIV analog Nle1-AngIV (Fig. 2), facilitated novel object recognition and spatial memory (Lee et al., 2004; Olson and Cero, 2010).

View all citing articles on Scopus
View full text