Elsevier

Pharmacology & Therapeutics

Volume 153, September 2015, Pages 36-54
Pharmacology & Therapeutics

Neuroprotective peptides fused to arginine-rich cell penetrating peptides: Neuroprotective mechanism likely mediated by peptide endocytic properties

https://doi.org/10.1016/j.pharmthera.2015.06.002Get rights and content

Abstract

Several recent studies have demonstrated that TAT and other arginine-rich cell penetrating peptides (CPPs) have intrinsic neuroprotective properties in their own right. Examples, we have demonstrated that in addition to TAT, poly-arginine peptides (R8 to R18; containing 8–18 arginine residues) as well as some other arginine-rich peptides are neuroprotective in vitro (in neurons exposed to glutamic acid excitotoxicity and oxygen glucose deprivation) and in the case of R9 in vivo (after permanent middle cerebral artery occlusion in the rat). Based on several lines of evidence, we propose that this neuroprotection is related to the peptide's endocytosis-inducing properties, with peptide charge and arginine residues being critical factors. Specifically, we propose that during peptide endocytosis neuronal cell surface structures such as ion channels and transporters are internalised, thereby reducing calcium influx associated with excitotoxicity and other receptor-mediated neurodamaging signalling pathways. We also hypothesise that a peptide cargo can act synergistically with TAT and other arginine-rich CPPs due to potentiation of the CPPs endocytic traits rather than by the cargo-peptide acting directly on its supposedly intended intracellular target. In this review, we systematically consider a number of studies that have used CPPs to deliver neuroprotective peptides to the central nervous system (CNS) following stroke and other neurological disorders. Consequently, we critically review evidence that supports our hypothesis that neuroprotection is mediated by carrier peptide endocytosis. In conclusion, we believe that there are strong grounds to regard arginine-rich peptides as a new class of neuroprotective molecules for the treatment of a range of neurological disorders.

Introduction

In recent years there has been an increased interest in the use of specifically designed peptides targeting cyto-damaging or cyto-protective pathways as neuroprotective agents. There are several reasons why this interest arose, including: i) peptide sequences critical for neurodamaging or neuroprotective intracellular protein–protein interactions can be easily identified and used as competitive inhibitors of target proteins (e.g. JNKI-1 peptide); ii) small peptides (2–40 amino acids) can be synthesised relatively cheaply using commercial sources; and iii) the development of cell penetrating peptides (CPPs), also referred to as protein or peptide transduction domains (PTDs), has provided a way to deliver peptides and other cargos (incl. proteins, nucleic acids and drugs) into cells and across the blood–brain barrier.

The discovery of CPPs has led to studies on the ability of a number of peptides and proteins to act as neuroprotection agents, as well as providing a means to explore the role of protein/protein interactions in brain function in health and disease (viz. neurological and non-neurological disorders). The main focus of this review is the use of arginine-rich CPPs (mainly TAT) for the delivery of neuroprotective peptides (<40 amino acids) particularly in cerebral ischaemia and stroke. The recent observation that CPPs have intrinsic neuroprotective properties in their own right has led us to question the conclusions of other studies. Here, we critically reappraise previous studies that have used putative neuroprotective peptides fused to CPPs as agents in cerebral ischaemia and other models of CNS injury, and examine the mechanism whereby arginine rich-peptides exert their neuroprotective effects. Importantly, we highlight that many past studies on neuroprotective peptides that have used cationic CPPs for CNS delivery may need to be reinterpreted in the light of the intrinsic neuroprotective effects of the carrier-peptide.

Cell penetrating peptides (CPPs) are small peptides (typically 5–25 amino acids) that are commonly used to facilitate the delivery of normally non-permeable cargo molecules such as other peptides, proteins, nucleic acids or drugs into cells, and across the blood–brain barrier. The development of CPPs as drug vehicles was sparked by the discovery of the PTD within the human immunodeficiency virus-type 1 trans-activator of transcription (HIV-TAT) protein (Frankel and Pabo, 1988, Green and Loewenstein, 1988). The active transporting peptide sequence within the HIV-TAT protein was isolated (TAT48–57: GRKKRRQRRR) and is now referred to as the TAT peptide or TAT (Becker-Hapak et al., 2001). Subsequently, over 100 CPPs have been identified (Milletti, 2012).

By far the most commonly used CPP peptide is TAT, especially to deliver various cargo molecules to the brain, including neuroprotective peptides and proteins. Other CPPs include penetratin (also known as antennapedia), poly-arginine peptides (R8 to R12; where R refers to arginine residues), Pep-1 and transportan. The amino acid sequences for these peptides, as well as of some less commonly used CPPs, are shown in Table 1. TAT, poly-arginine and penetratin are cationic arginine-rich CPPs.

Potential neuroprotective peptides fused to CPPs have been assessed in cultured neurons and animal models that mimic neural injury mechanisms seen in a variety of disorders, including cerebral ischaemia, spinal cord injury, traumatic brain injury, epilepsy, Parkinson's disease and Alzheimer's disease (Arthur et al., 2007, Colombo et al., 2007, Lai et al., 2005, Liu et al., 2006, Meade et al., 2009, Nagel et al., 2008). However, several years ago, we and others demonstrated that TAT possesses intrinsic neuroprotective properties both in vitro in neurons exposed to excitotoxicity and oxygen–glucose deprivation (OGD) and in vivo following cerebral ischaemia in P12 rats after intraventricular injection (Craig et al., 2011, Meade et al., 2010a, Vaslin et al., 2009b, Xu et al., 2008). We subsequently showed that poly-arginine-9 (R9), penetratin and Pep-1 also display neuroprotective actions in in vitro excitotoxic and/or OGD models (Meloni et al., 2014). Furthermore, our data showed that R9 and penetratin were 17- and 4.6-fold respectively more neuroprotective than TAT (Meloni et al., 2014).

The higher potency of R9 relative to TAT and penetratin led us to explore the in vitro neuroprotective potency of other poly-arginine peptides (R1, R3, R6–R15 and R18), as well as, other arginine-rich peptides (Meloni et al., 2015). These studies confirmed that poly-arginine and arginine-rich peptides as a group are highly neuroprotective, with efficacy increasing with increasing arginine content, peaking at R15 (Meloni et al., 2015). We also showed that arginine-rich peptides have the capacity to reduce glutamic acid-induced neuronal calcium influx and are neuroprotective with a single treatment several hours before glutamic acid or OGD exposure. Furthermore, neuroprotective efficacy was shown to be directly related to peptide positive net charge conferred by the positively charged arginine (R) and lysine (K) amino acids residues, which could be blocked by fusion with a negatively charged glutamic acid (E9) poly-peptide (e.g. R9/E9 peptide) or by incubation with the highly negatively charged molecule heparin. The latter finding strongly suggests that peptides bind to negatively charged cell surface molecules such as heparin sulphate proteoglycans (HSPGs), chondroitin sulfate proteoglycans (CSPGs) or sialic acid residues present in glycosphingolipids to initiate and stimulate peptide endocytosis (Favretto et al., 2014, Kim et al., 2012, Ravindran et al., 2013, Wallbrecher et al., 2014) a process crucial for neuroprotection (Meloni et al., 2015). In this context, others have demonstrated that the nature of the peptide interaction with HSPGs determines CPPs endocytic properties (Wallbrecher et al., 2014).

With respect to endocytosis, studies have demonstrated that peptide charge conferred by arginine and lysine residues (note: arginine and lysine are the only two strongly positively charged amino acids, with histidine being only weakly positively charged, whereas glutamic acid and aspartic acid are the only two negatively charged amino acids) facilitate HSPG binding, and that mainly arginine residues trigger the endocytic process (Amand et al., 2012, Wallbrecher et al., 2014, Yang et al., 2014). Consistent with our proposed endocytic neuroprotective mechanism, we have demonstrated that poly-lysine (K10) is only weakly neuroprotective in a cortical neuronal glutamic acid excitotoxicity model (Meloni et al., 2015). It is also likely that other amino acids can influence the endocytic properties of cationic peptides in both a positive and negative manner as has been demonstrated for tryptophan (W; Rydberg et al., 2012, Bechara et al., 2013) and alanine (A; Yang et al., 2014), respectively. Indeed, we have now confirmed that tryptophan and alanine amino acids within arginine-rich peptides respectively increase and decrease neuroprotective efficacy in a glutamic acid excitotoxicity model (Fig. 1).

Based on our recent findings we hypothesised that arginine-rich peptides exert their neuroprotection effects by inducing the endocytic internalisation of cell surface ion channels, thereby reducing the damaging effects of excitotoxicity (see Fig. 2). This is a novel hypothesis that essentially identifies arginine-rich peptides as a new class of neuroprotective molecule. There are several lines of evidence based on our findings and those of others that support our endocytosis hypothesis. Arginine-rich peptides, including so called “neuroprotective peptides” fused to TAT have been shown to: i) reduce neuronal calcium influx (Meloni et al., 2015) and interfere with ion channel function (NMDA receptor: Ferrer-Montiel et al., 1988, Tu et al., 2010, Sinai et al., 2010, Brittain et al., 2011b, Brustovetsky et al., 2014, VR1: Planells-Cases et al., 2000, CaV2.2: Brittain et al., 2011a, Brittain et al., 2011b, Feldman and Khanna, 2013, Brustovetsky et al., 2014; sodium calcium exchanger [NCX], CaV3.3: García-Caballero et al., 2014); ii) cause internalisation of neuronal ion channels (Brustovetsky et al., 2014, Sinai et al., 2010); and iii) require endocytosis as a prerequisite for neuroprotection (Meloni et al., 2015, Vaslin et al., 2011). Interestingly, other TAT-fused peptides have also been shown to interfere with the function of neuronal receptors (D1R–D2R; Pei et al., 2010; PTPσ: Lang et al., 2015). In this context, it is important to note that endocytosis is a known mechanism used by cells to internalise cell surface receptors (Höller and Dikic, 2004, Marchese, 2014, Maxfield and McGraw, 2004).

Neuroprotective efficacy, at least for poly-arginine peptides (Meloni et al., 2015), appears to correlate with peptide transduction efficacy (Mitchell et al., 2000), a process known to occur by endocytosis (Appelbaum et al., 2012, Bechara et al., 2013, El-Sayed et al., 2009). Furthermore, it is important to note that the rapid and transient (lasting up to 4 h with peptide pre-treatment) nature of the neuroprotection induced by poly-arginine peptides (Meloni et al., 2015) corresponds closely to the timeframes of endocytosis and endosomal receptor re-cycling (Gundelfinger et al., 2003, Maxfield and McGraw, 2004, Yashunsky et al., 2009). Importantly, it is known that TAT, penetratin and R9 can induce the internalisation of EGFR and TNFR in HeLa cells (Fotin-Mleczek et al., 2005). Our hypothesis also links endocytosis as a common neuroprotective mechanism of action for a diverse range of arginine-rich peptides (including TAT-fused peptides), all of which are likely to have endocytic inducing properties.

This neuroprotective mechanism that we propose is also consistent with the link between neuronal cell surface-HSPGs (Litwack et al., 1994) and endocytic activity (Vaslin et al., 2009a), which are known to promote endosomal uptake of cationic CPPs (Nakase et al., 2007, Vaslin et al., 2009a, Vaslin et al., 2011). It is also possible that other negatively charged cell surface receptors such as CSPGs and glycosphingolipids can promote cationic CPP endocytosis and neuroprotection. As mentioned above, positively charged poly-arginine and arginine-rich peptides are known to bind negatively charged HSPGs to initiate endocytosis. It is important to note that any neuroprotective peptide fused to a CPP and internalised by endocytosis must escape the endosome to interact with its intended cytoplasmic target. However, endosomal escape appears to be a highly inefficient process (Appelbaum et al., 2012, Qian et al., 2014) (and rarely confirmed) and as a result, due to the cargo's inability to engage with its intracellular target it is unlikely to have a significant impact within the cytoplasm.

In light of our recent findings, the aim of this review is to critically re-examine studies in the literature that have used neuroprotective peptides fused to cationic CPPs (i.e. TAT, R9) and present evidence supporting our hypothesis that the neuroprotective actions of these peptides are primarily, if not exclusively, due to the endocytic properties of the peptide per se.

Section snippets

Examination of studies using CPP-fused to neuroprotective peptides in neuronal injury models

To date, over a dozen of neuroprotective peptides fused to CPPs have been described (Table 2, Table 3, Table 4). Three of the most intensely studied peptides developed as potential neuroprotective agents for stroke/cerebral ischaemia are NR2B9c, JNKI-1 and CBD3 (Table 2, Table 3, Table 4). This review in particular critically examines the use of these three peptides in a neurological setting, and provides evidence suggesting that the critical neuroprotective and functional structural elements

Examination neuroprotective arginine- (and lysine-) rich peptides used in neuronal injury models

It is beyond the scope of this review to include every study that has used a neuroprotective TAT-fused peptide. For completeness, other in vivo studies using TAT-fused peptides are listed in Table 5. Evidently, several studies have identified arginine- and lysine-rich peptides not fused to TAT or a CPP as being neuroprotective. We argue that it is possible that these peptides are intrinsically neuroprotective via mechanisms unrelated to their proposed action on a specific cell surface receptor

Discussion and concluding remarks

The main purpose of this review is to describe the neuroprotective properties of peptides fused to the arginine-rich CPP, TAT and in doing so provide evidence supportive of our hypothesis that neuroprotection is mediated not by the cargo molecule but largely by TAT itself. This hypothesis crystallised for us following the analysis of the neuroprotective and calcium influx inhibiting properties of a diverse set of peptides including: 1) arginine-rich CPPs (TAT, penetratin); 2) poly-arginine

Conflict of interest

B.P. Meloni and N.W. Knuckey are named inventors of several patent applications regarding the use of arginine-rich peptides as neuroprotective agents. The other authors declare no conflict of interest.

Acknowledgments

This study in part was supported by the Department of Neurosurgery, Sir Charles Gairdner Hospital and by a Neurotrauma Research Program of Western Australia research grant.

References (140)

  • J.W. Chambers et al.

    Blocking c-Jun N-terminal kinase (JNK) translocation to the mitochondria prevents 6-hydroxydopamine-induced toxicity in vitro and in vivo

    J Biol Chem

    (2013)
  • F. Corrigan et al.

    The neuroprotective domains of the amyloid precursor protein, in traumatic brain injury, are located in the two growth factor domains

    Brain Res

    (2011)
  • A. El-Sayed et al.

    Enhanced gene expression by a novel stearylated INF7 peptide derivative through fusion independent endosomal escape

    J Control Release

    (2009)
  • E. Esneault et al.

    d-JNKi, a peptide inhibitor of c-Jun N-terminal kinase, promotes functional recovery after transient focal cerebral ischemia in rats

    Neuroscience

    (2008)
  • M.E. Favretto et al.

    Glycosaminoglycans in the cellular uptake of drug delivery vectors — Bystanders or active players?

    J Control Release

    (2014)
  • P. Feldman et al.

    Challenging the catechism of therapeutics for chronic neuropathic pain: Targeting CaV2.2 interactions with CRMP2 peptides

    Neurosci Lett

    (2013)
  • A.D. Frankel et al.

    Cellular uptake of the tat protein from human immunodeficiency virus

    Cell

    (1988)
  • A. García-Caballero et al.

    The deubiquitinating enzyme USP5 modulates neuropathic and inflammatory pain by enhancing Cav3.2 channel activity

    Neuron

    (2014)
  • D.J. Goebel

    Selective blockade of CaMKII-alpha inhibits NMDA-induced caspase-3-dependent cell death but does not arrest PARP-1 activation or loss of plasma membrane selectivity in rat retinal neurons

    Brain Res

    (2009)
  • M. Green et al.

    Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein

    Cell

    (1988)
  • Q.H. Guan et al.

    Neuroprotection against ischemic brain injury by a small peptide inhibitor of c-Jun N-terminal kinase (JNK) via nuclear and non-nuclear pathways

    Neuroscience

    (2006)
  • M.D. Hill et al.

    Safety and efficacy of NA-1 in patients with iatrogenic stroke after endovascular aneurysm repair (ENACT): A phase 2, randomised, double-blind, placebo-controlled trial

    Lancet Neurol

    (2012)
  • D. Höller et al.

    Receptor endocytosis via ubiquitin-dependent and -independent pathways

    Biochem Pharmacol

    (2004)
  • A. Ishida et al.

    A novel highly specific and potent inhibitor of calmodulin-dependent protein kinase II

    Biochem Biophys Res Commun

    (1995)
  • T.K. Kao et al.

    Opioids modulate post-ischemic progression in a rat model of stroke

    Neurochem Int

    (2008)
  • N.A. Kaufman et al.

    COG1410, an apolipoprotein E-based peptide, improves cognitive performance and reduces cortical loss following moderate fluid percussion injury in the rat

    Behav Brain Res

    (2010)
  • X.M. Liu et al.

    Neuroprotection of Tat-GluR6-9c against neuronal death induced by kainate in rat hippocampus via nuclear and non-nuclear pathways

    J Biol Chem

    (2006)
  • Z. Liu et al.

    CaMKII antisense oligodeoxynucleotides protect against ischemia-induced neuronal death in the rat hippocampus

    J Neurol Sci

    (2012)
  • C.F. Lourenço et al.

    Neurovascular coupling in hippocampus is mediated via diffusion by neuronal-derived nitric oxide

    Free Radic Biol Med

    (2014)
  • M. Lundberg et al.

    Cell surface adherence and endocytosis of protein transduction domains

    Mol Ther

    (2003)
  • J.R. Lynch et al.

    APOE genotype and an ApoE-mimetic peptide modify the systemic and central nervous system inflammatory response

    J Biol Chem

    (2003)
  • J.R. Lynch et al.

    A novel therapeutic derived from apolipoprotein E reduces brain inflammation and improves outcome after closed head injury

    Exp Neurol

    (2005)
  • A. Marchese

    Endocytic trafficking of chemokine receptors

    Curr Opin Cell Biol

    (2014)
  • J.D. McAdoo et al.

    Intrathecal administration of a novel apoE-derived therapeutic peptide improves outcome following perinatal hypoxic–ischemic injury

    Neurosci Lett

    (2005)
  • A.J. Meade et al.

    AP-1 inhibitory peptides attenuate in vitro cortical neuronal cell death induced by kainic acid

    Brain Res

    (2010)
  • F. Milletti

    Cell-penetrating peptides: Classes, origin, and current landscape

    Drug Discov Today

    (2012)
  • T. Nakazawa et al.

    Characterization of Fyn-mediated tyrosine phosphorylation sites on GluR epsilon 2 (NR2B) subunit of the N-methyl-d-aspartate receptor

    J Biol Chem

    (2001)
  • C.H. Nijboer et al.

    Mitochondrial JNK phosphorylation as a novel therapeutic target to inhibit neuroinflammation and apoptosis after neonatal ischemic brain damage

    Neurobiol Dis

    (2013)
  • C.H. Nijboer et al.

    Inhibition of the JNK/AP-1 pathway reduces neuronal death and improves behavioral outcome after neonatal hypoxic–ischemic brain injury

    Brain Behav Immun

    (2010)
  • M. Aarts et al.

    Treatment of ischemic brain damage by perturbing NMDA receptor–PSD-95 protein interactions

    Science

    (2002)
  • T. Abe et al.

    Fyn kinase-mediated phosphorylation of NMDA receptor NR2B subunit at Tyr1472 is essential for maintenance of neuropathic pain

    Eur J Neurosci

    (2005)
  • P.G. Arthur et al.

    Necrotic death of neurons following an excitotoxic insult is prevented by a peptide inhibitor of c-jun N-terminal kinase

    J Neurochem

    (2007)
  • A. Bach et al.

    A high-affinity, dimeric inhibitor of PSD-95 bivalently interacts with PDZ1-2 and protects against ischemic brain damage

    Proc Natl Acad Sci U S A

    (2012)
  • F.C. Barone et al.

    Inhibition of p38 mitogen-activated protein kinase provides neuroprotection in cerebral focal ischemia

    Med Res Rev

    (2001)
  • C. Bechara et al.

    Tryptophan within basic peptide sequences triggers glycosaminoglycan-dependent endocytosis

    FASEB Journal

    (2013)
  • K.F. Bell et al.

    Calmodulin kinase IV-dependent CREB activation is required for neuroprotection via NMDA receptor-PSD95 disruption

    J Neurochem

    (2013)
  • A.C. Bessero et al.

    Role of the c-Jun N-terminal kinase pathway in retinal excitotoxicity, and neuroprotection by its inhibition

    J Neurochem

    (2010)
  • T. Borsello et al.

    A peptide inhibitor of c-Jun N-terminal kinase protects against excitotoxicity and cerebral ischemia

    Nat Med

    (2003)
  • B.T. Bråtane et al.

    Neuroprotection by freezing ischemic penumbra evolution without cerebral blood flow augmentation with a postsynaptic density-95 protein inhibitor

    Stroke

    (2011)
  • R. Bright et al.

    Protein kinase C delta mediates cerebral reperfusion injury in vivo

    J Neurosci

    (2004)
  • Cited by (71)

    • The therapeutic potential of the neuroactive peptides of soluble amyloid precursor protein-alpha in Alzheimer's disease and related neurological disorders

      2023, Seminars in Cell and Developmental Biology
      Citation Excerpt :

      HSPG-mediated peptide endocytosis is initiated by CARPs binding to HSPGs [103,104]. Therefore, APP96–110 peptide binding may lead to neuroprotection by preventing neuronal excitotoxicity, via endocytosis of cell surface receptors, such as the NMDA receptor subunit NR2B, a key mediator of neuronal excitotoxic calcium influx [104–106]. Interestingly, although RERMS has a charge of only + 1 at pH 7, it fulfils the remaining CARP definition requirements and so potentially may mediate, in part, its effects via these mechanisms also.

    • Assessment of the safety of the cationic arginine-rich peptides (CARPs) poly-arginine-18 (R18 and R18D) in ex vivo models of mast cell degranulation and red blood cell hemolysis

      2022, Biochemistry and Biophysics Reports
      Citation Excerpt :

      It also includes the arginine-rich peptide protamine, which was approved by the United States Food and Drug Administration (FDA) as a reversal agent for the anticoagulant heparin in 1939 and as a constituent in some insulin preparations to prolong euglycaemia in 1950. The R18 and R18D peptides are lead neuroprotective molecules in our laboratory and have demonstrated beneficial effects in rat models of ischemic stroke, hypoxic-ischemic encephalopathy and traumatic brain injury [2–11] and in a non-human primate stroke model [12]. Given the potential future application of R18 or R18D as an ischemic stroke neurotherapeutic, it is important to establish their capacity to cause undesirable side effects such as mast cell induced anaphylactoid reactions and red blood cell (RBC) hemolysis when administered to patients.

    View all citing articles on Scopus
    View full text