Next Article in Journal
The Expanding Role of Ketogenic Diets in Adult Neurological Disorders
Next Article in Special Issue
Haemodynamic Instability and Brain Injury in Neonates Exposed to Hypoxia–Ischaemia
Previous Article in Journal
Reading Deficits in Intellectual Disability Are still an Open Question: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Perinatal Hypoxic-Ischemic Encephalopathy and Neuroprotective Peptide Therapies: A Case for Cationic Arginine-Rich Peptides (CARPs)

by
Adam B. Edwards
1,2,3,*,
Ryan S. Anderton
1,2,4,
Neville W. Knuckey
1,3,4 and
Bruno P. Meloni
1,3,4
1
Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
2
School of Health Sciences and Institute for Health Research, The University of Notre Dame Australia, Fremantle, WA 6160, Australia
3
Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA 6009, Australia
4
Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
*
Author to whom correspondence should be addressed.
Brain Sci. 2018, 8(8), 147; https://doi.org/10.3390/brainsci8080147
Submission received: 28 June 2018 / Revised: 25 July 2018 / Accepted: 1 August 2018 / Published: 7 August 2018
(This article belongs to the Special Issue The Treatment of Neonatal Hypoxic-Ischemic Encephalopathy)

Abstract

:
Perinatal hypoxic-ischemic encephalopathy (HIE) is the leading cause of mortality and morbidity in neonates, with survivors suffering significant neurological sequelae including cerebral palsy, epilepsy, intellectual disability and autism spectrum disorders. While hypothermia is used clinically to reduce neurological injury following HIE, it is only used for term infants (>36 weeks gestation) in tertiary hospitals and improves outcomes in only 30% of patients. For these reasons, a more effective and easily administrable pharmacological therapeutic agent, that can be used in combination with hypothermia or alone when hypothermia cannot be applied, is urgently needed to treat pre-term (≤36 weeks gestation) and term infants suffering HIE. Several recent studies have demonstrated that cationic arginine-rich peptides (CARPs), which include many cell-penetrating peptides [CPPs; e.g., transactivator of transcription (TAT) and poly-arginine-9 (R9; 9-mer of arginine)], possess intrinsic neuroprotective properties. For example, we have demonstrated that poly-arginine-18 (R18; 18-mer of arginine) and its D-enantiomer (R18D) are neuroprotective in vitro following neuronal excitotoxicity, and in vivo following perinatal hypoxia-ischemia (HI). In this paper, we review studies that have used CARPs and other peptides, including putative neuroprotective peptides fused to TAT, in animal models of perinatal HIE. We critically evaluate the evidence that supports our hypothesis that CARP neuroprotection is mediated by peptide arginine content and positive charge and that CARPs represent a novel potential therapeutic for HIE.

1. Introduction

Perinatal hypoxic-ischemic encephalopathy (HIE; also referred to as birth asphyxia) remains the leading cause of neonatal mortality and morbidity, with an incidence in developed nations of 2–6 and 7 in every 1000 live term (>36 weeks gestation) and pre-term (≤36 weeks gestation) births, respectively [1,2]. The incidence is even higher in developing countries, with rates of up to 30 per 1000 live full-term births [3,4]. Of the individuals affected by HIE, 15–20% will die in the postnatal period, and an additional 25% will develop severe and permanent neurological sequela. Each year, HIE accounts for up to 1.2 million deaths [5] and some form of central nervous system (CNS) dysfunction in an estimated 1.15 million neonates [6] such as cerebral palsy, epilepsy, global developmental delay, intellectual disability, behavioral disorders and autism spectrum disorders [7,8,9,10,11]. While hypothermia is used as a therapeutic intervention to minimize brain injury in neonates suffering HIE, it only provides beneficial outcomes in approximately 30% of patients and has several limitations (reviewed below). Therefore, an effective and easily administrable pharmacological therapeutic agent to improve patient outcomes when used as an adjunct to hypothermia, on its own or when hypothermia cannot be applied is urgently needed.
In recent years, there has been an increased application of putative neuroprotective peptides designed to target cyto-damaging or cyto-protective pathways to reduce injury in acute brain disorders such as HIE. One reason for this interest is the discovery of cell-penetrating peptides (CPPs), allowing for the delivery of neuroprotective peptides and other molecular cargos (e.g., proteins, nucleic acids and nanoparticles) across the blood brain barrier (BBB) and into cells within the CNS. Most studies assessing neuroprotective peptides in animal models of HIE use the cationic arginine-rich CPP, transactivator of transcription (TAT47–57 YGRKKRRQRRR), as the ‘carrier’ molecule. However, several years ago, our and other laboratories demonstrated that the TAT peptide has modest intrinsic neuroprotective properties in its own right [12,13,14,15,16,17]. We subsequently demonstrated that other cationic arginine-rich peptides [CARPs; e.g., penetratin, protamine, sodium calcium exchanger inhibitory peptide (XIP), poly-arginine peptides (e.g., R9, R12 and R18; R indicates amino acid arginine and number indicates mer)], possess potent neuroprotective properties in both in vitro and animal models of cerebral ischemia and/or hypoxia [12,18,19,20,21,22,23,24,25,26,27,28,29], and demonstrated that the arginine content and peptide positive charge are critical determinants of neuroprotection [19,27,28].
Based on the recent application of cationic CPPs for the delivery of putative neuroprotective peptides in animal models of HIE and the recent discovery of the intrinsic neuroprotective properties of CARPs, the objectives of this review are: (i) catalogue studies that have used peptides as neuroprotective agents in HIE, especially those employing a CPP delivery system; and (ii) to evaluate the potential neuroprotective effects of both CPP and cargo molecules in terms of their arginine content and cationic charge, as well as the content of other potentially neuroprotective amino acids such as tryptophan [29]. We will propose that the neuroprotective properties of most, if not all putative neuroprotective peptides fused to cationic CPPs are likely to be mediated in a large part by the actions of the carrier CPP (e.g., TAT) rather than the cargo molecule itself, with potency being further enhanced according to the amino acid content of the cargo (e.g., number of arginine residues) [19]. In addition, we will also review the evidence that CARPs have multiple cellular modes of action that underlie their neuroprotective properties.
First, we provide a brief overview of the major underlying pathophysiological mechanisms associated with HIE and the application of hypothermia as an acute therapy for the disorder.

2. Pathophysiology of Perinatal Hypoxia-Ischemic Brain Injury

2.1. Initiation of the Pathophysiological Cascade in HIE

The etiology of HIE is associated with a variety of maternal, placental and fetal conditions; all of which have variable clinical manifestations. These conditions include, but are not limited to, chronic maternal hypoxia (e.g., chronic utero-placental hypoxia, cardiopulmonary arrest, acute hypotension, pulmonary embolism or vascular disease), pre-eclampsia, nuchal cord, umbilical cord knotting, umbilical cord prolapse, shoulder dystocia and premature placental detachment [30]. Perinatal hypoxia is initiated following an impairment of oxygenated cerebral blood flow (CBF) to the fetus, triggering responses at both the systemic and cellular levels. The fetal brain is particularly sensitive to a HI environment, as there is a requirement of a constant supply of energy in the form of ATP, and when interrupted, excitotoxicity occurs through the uncontrolled release of excitatory neurotransmitters such as glutamate; marking the beginning of the ischemic cascade. The acute and downstream consequences of the ischemic cascade are damaging to neurons and other cells (e.g., glial progenitor cells and astrocytes) at the cytoplasmic and mitochondrial levels, as well as causing disruption to the BBB and the activation of inflammatory responses.

2.2. Excitotoxicity

The metabolic rate of the fetal brain is largely determined by the stage of gestational development [30]. To meet metabolic demands, developing neural tissue metabolizes lactate, ketone bodies and glucose. When compared to the developed brain, the fetal brain is more adaptable with respect to energy utilization, thereby increasing its capacity to tolerate hypoxia-ischemia (HI) [31]. However, in the event of a critical depletion of ATP due to prolonged HI, the fetal brain eventually becomes susceptible to injury.
Following prolonged HI, cellular homeostasis is disrupted due to the depletion of ATP and failure to maintain ionic gradients, and as a result, neurons depolarize and release glutamate into the synaptic cleft [32]. High extracellular levels of glutamate cause excitotoxicity in neurons and other cells, namely glial progenitor cells, that express glutamate receptors (NMDA; N-methyl-d-asparate, AMPA; α-amino-3-hydroxy-5-methy-4-isoxazolepropionic acid, kainic acid and metabotropic glutamate receptors), producing a cytotoxic cellular influx of calcium ions [33]. The increased levels of intracellular calcium in neurons and glial progenitor cells in turn results in the activation of calcium-dependent proteases, lipases and deoxyribonuclease (DNase), reactive oxygen species (ROS) production, oxidative stress, cytotoxic edema, mitochondrial dysfunction and the stimulation of pro-cell death pathways [34,35,36,37,38].

2.3. Oxidative Stress

Oxidative stress plays an important role in the pathophysiology of HIE, as the developing brain comprises high levels of unsaturated fatty acids susceptible to lipid peroxidation, metals catalyzing free radical reactions and low concentrations of antioxidants [39]. The resulting heightened sensitivity to oxidative stress and ROS production following HI causes significant damage to lipids (lipid oxidation), nucleic acids (DNA degeneration) and proteins (protein oxidation). Sources of ROS following HI include, the mitochondrial electron transport chain (ETC), NADPH oxidases (NOX), xanthine oxidase (XO), arachidonic acid (12/15 lipoxygenase), and nitric oxide synthase (NOS). In particular, NOS has been targeted in HIE, with strategies to ablate or inhibit NOS activity invariably being neuroprotective in animal models of HIE [40].

2.4. Mitochondrial Dysfunction

Mitochondria have diverse functions, including ATP generation, intracellular calcium regulation, ROS production, biosynthesis of amino acids, lipids and nucleotides and pro-cell death signaling. The role of mitochondria in HIE has been extensively reviewed [41,42,43]. Here we focus on the role of mitochondria in potentiating secondary pathophysiological cascades contributing to HI. Mitochondria contribute to secondary brain injury following HI via the regulation of cell death pathways, namely apoptosis. Cellular apoptosis following HI is activated via both intrinsic (e.g., in response to mitochondrial dysfunction and damaged DNA) and extrinsic [e.g., in response to cell death receptors such as Fas and tissue necrosis factor-α (TNF-α)] pathways and involve the release of pro-cell death proteins from mitochondria.
In cells affected by HI, several processes such as excitotoxicity, ROS production, activation of p53, DNA damage and altered mitochondrial function can influence the activity of pro-apoptotic Bcl-2 proteins (e.g., Bid and Bax), resulting in Bax-dependent mitochondrial membrane permeability transition (MPT) [44,45]. In addition, excessive mitochondrial ROS generation, due to leakage from the ETC, can oxidize the inner mitochondrial membrane phospholipid, cardiolipin, promoting mitochondrial membrane permeability transition pore (MPTP) opening and subsequent release of cytochrome-c. Cytosolic cytochrome-c, along with deoxyadenosine triphosphate (dATP), interact with apoptotic protease activating factor-1 (Apaf-1) to form the apoptosome, which cleaves and activates pro-caspase-9 [46]. Activated caspase-9 in turn activates caspase-3, which is a major proteolytic enzyme responsible for the ultimate dismantling of cells during apoptotic cell death.
Further alterations to mitochondria, such as mitochondrial outer membrane permeabilization (MOMP), results in the release other pro-apoptotic factors into the cytosol, including apoptosis-inducing factor (AIF), endonuclease G (Endo-G), and Smac/DIABLO [47,48]. Translocation of AIF and Endo-G to the nucleus mediates chromatin fragmentation and Smac/DIABLO interacts with inhibitors of apoptosis to reduce their actions on activated caspases.
TNF cell death receptor signaling mainly stimulates the extrinsic pathway by activating caspase-8, which activates other caspases, including caspase-3. While beyond the scope of this review, it is also important to note that other cell death pathways associated with necrosis, necroptosis, ferroptosis and autophagy may also be activated in response to HI. These pathways can be activated in response to one or more cellular disturbances such as calcium influx, mitochondrial dysfunction, cellular energy depletion, ROS production, TNF receptors cell signaling and other cellular cell signaling pathways (e.g., adenosine monophosphate-activated protein kinase).

2.5. Inflammation

The inflammatory response following HIE has been extensively reviewed by [49,50] and is therefore briefly discussed. In the immature brain following HI, an innate immune response occurs within minutes of the injury [51]. Initially, microglia become activated within the cerebral parenchyma, followed by a systemic inflammatory response, mediated by the infiltration of circulating monocytes, neutrophils and T-cells into the brain. Activated microglia develop macrophage-like attributes, such as phagocytic properties, cytokine production, antigen presentation and matrix metalloproteinase (MMP) release. The release and activation of MMPs by microglia can compromise BBB integrity, further promoting the invasion of peripheral leukocytes into the cerebral parenchyma; exacerbating ensuing cerebral injury. A hallmark of HIE is the aggregation of amoeboid microglia within the periventricular white matter, resulting in the production of inflammatory cytokines (TNF-α and IL1-β), NO and other ROS [52]; collectively potentiating the toxic effects of the ischemic cascade in neurons, glial progenitor cells and the cerebral vasculature.
While astrocytes attempt to provide neuroprotective support to neurons and glia via the release of glutathione, superoxide dismutase (SOD), extra-synaptic uptake of glutamate and the maintenance of ion channel gradients, these protective mechanisms can be quickly overrun. As with microglia, astrocytes become hyper-activated due to the effects of pro-inflammatory cytokines, ROS, and dying neurons and glia. Activated astrocytes also release pro-inflammatory cytokines (IL-6, TNF-α, IL-1α/β and INF-γ), further exacerbating HI-induced cell death pathways (e.g., apoptosis). In addition, astrocytes secrete chemokines that attracts systemic immune cells to infiltrate affected tissue, further aggravating tissue injury.

3. Current Clinical Treatments: Hypothermia

Hypothermia is considered one of the most effective neuroprotective treatment interventions in both experimental and clinical settings. While the exact beneficial effects of hypothermia are yet to be elucidated in HIE, it is known to suppress many of the neuro-damaging events associated with HI including excitotoxicity, apoptosis, inflammation, oxidative stress and BBB disruption [53].
In HIE, moderate hypothermia (33.5 °C for 72 h), applied within 6 h of birth, improves the survival and neurodevelopmental outcomes in infants suffering from moderate and severe HIE. While hypothermia is now considered standard care in the treatment of HIE and is well tolerated in term infants [54,55,56,57], it does not provide complete protection. Of the infants who have moderate or severe encephalopathy, therapeutic hypothermia was shown to decrease mortality from 40% to 28%, while in surviving infants neurological morbidity was reduced from 31% to 19% of infants receiving therapeutic hypothermia [58].
Importantly, while hypothermia is safe in pre-term infants with necrotising enterocolitis [59], it has not been adequately evaluated for use in pre-term infants suffering HIE. Information regarding the safety and efficacy of hypothermia to treat pre-term infants suffering HIE is lacking, although information may be available in the near future (NCT1793129). In one study, hypothermia in pre-term infants was associated with higher mortality and increased clinical complications when compared to term infants [60], although the study did not include a normothermic pre-term control group. The inadequate application of hypothermia in the pre-term neonate highlights the need for the development of safer therapeutic interventions in these patients following HI.
In addition, the induction of hypothermia requires specialized equipment, intensive care monitoring and trained staff, limiting its use to centers that have the facilities and medical personnel to undertake the procedure. This develops uncertainty regarding the timing, safety and efficacy of initiating hypothermia in infants before transfer to a centre equipped to deliver hypothermia. For example, the transfer of infants suffering HIE to a medical centre to provide hypothermia can exceed 6 h from the time of birth and that attempts of passive cooling during transport can result in excessive cooling [61]. In light of this, there is an urgent need for the application of an acute neuroprotective therapy for infants who suffer HIE when hypothermia cannot be applied immediately, or is not available. In addition, as the incidences of mortality and morbidity are still relatively high following hypothermia, there is a requirement for additional therapeutic agents, adjuvant to hypothermia, to further improve outcomes in infants suffering HIE.

4. Neuroprotective Peptides and Their Therapeutic Application in HIE

4.1. Peptide Therapeutics

Peptides represent an important and increasingly popular class of therapeutic agent. To date, over 60 general peptide drugs have been approved in the United States, Europe, and Japan, approximately 140 are in active clinical development and more than 500 peptides are being examined in preclinical therapeutic studies [62]. Peptides have several beneficial properties over traditional drugs such as high biological and structural diversity, less toxicity and accumulation in tissue and reduced propensity to cause drug-drug interactions. The main disadvantage of most peptides is their inability to cross the BBB and enter cells, which is an important issue if the peptide has an intracellular target and/or is targeting a CNS disorder. However, as outlined above, the delivery of peptides into the CNS and into cells can be achieved by using CPPs. For this reason, CPPs have been widely utilized as a carrier molecule for the delivery of neuroactive compounds into the CNS, including putative neuroprotective peptides. Below we review experimental studies that have used neuroprotective peptides in HIE models, including putative neuroprotective peptides fused to a CPP. First, we summarize findings from our own and other laboratories highlighting the intrinsic neuroprotective properties of CARPs.

4.2. CARPs Are Intrinsically Neuroprotective

An early study by Ferrer-Montiel et al. [63], demonstrated that cationic arginine-rich hexapeptides (i.e. containing 2–6 arginine residues; net charge +3–+7; e.g., RRRRRR-NH2) were neuroprotective in an in vitro hippocampal neuronal NMDA excitotoxic model. These findings were in line with subsequent studies in our and other laboratories, demonstrating that the CPP TAT (YGRKKRRQRRR; net charge +8) possesses intrinsic neuroprotective properties following in vitro glutamic acid excitotoxicity and oxygen glucose deprivation (OGD), as well as in vivo following excitotoxicity and cerebral ischemia in rats [13,14,15,16,64,65]. We subsequently demonstrated that in an in vitro cortical neuronal excitotoxic model, the CPPs poly-arginine-9 (RRRRRRRRR-NH2; net charge +10) and penetratin (RQIKIWFQNRRMKWKK-NH2; net charge +8) were even more potent than TAT [12]. The high potency of R9 led us to explore other poly-arginine peptides (e.g., R1, R3, R6–R15, R18, R22), as well as several other CARPs, including protamine in the in vitro excitotoxic and OGD neuronal injury models [27,28,29]. These studies confirmed that CARPs are highly neuroprotective, with efficacy increasing with arginine content and peptide positive charge; peaking at R15 to R18 for poly-arginine peptides [27,29] [Note arginine and lysine (K) are the only positively charged amino acids, while histidine has a low positive charge].
Our laboratory has also demonstrated that CARPs and in particular the poly-arginine peptide R18 are neuroprotective in in vivo experimental brain ischemic and/or hypoxic injury models [18,20,21,22,24,26,27,29], and that CARPs can reduce neuronal cell surface glutamate receptor levels and excitotoxic calcium influx [19,22,23,25,27,28]. As highlighted previously [19], based on our findings, we have proposed that the neuroprotective properties of most, if not all, putative neuroprotective peptides fused to cationic CPPs (e.g., TAT, R9 and penetratin), as well as other CARPs, are determined by the arginine content and positive charge of the peptide, with potency also influenced by other amino acid residues (e.g., lysine, tryptophan and phenylalanine) [19,29]. Importantly, an arginine and cationic charge mediated neuroprotective mechanism of action of CARPs, including CPP-fused to putative neuroprotective peptides (e.g., TAT-NR2B9c and TAT-NR2Bct), is supported in studies by Marshall et al. [65] and more recently McQueen et al. [66]. Based on studies from our own and other laboratories, we believe that the mechanism of action of putative neuroprotective peptides fused to a CPP need to be critically re-evaluated. In light of the potentially confounding effects of CPPs and/or peptide arginine content and positive charge, we will review studies using neuroprotective peptides in HIE models. First, we provide a summary of the known and potential neuroprotective actions of CARPs.

4.3. Cationic Arginine-Rich Cell-Penetrating Peptide Neuroprotective Mechanisms of Actions

While the exact mechanisms of the neuroprotective effects are still being explored, we and others have demonstrated that CARPs have multiple, potentially simultaneous mechanisms of action. CARPs and putative neuroprotective peptides fused to cationic arginine-rich CPPs (i.e., TAT or R9; e.g., JNKI-1-TATD, TAT-NR2B9c/NA-1, TAT-CBD3/R9-CBD3-A6K and TAT-3.2-III-IV) can mitigate excitotoxic or potassium evoked neuronal intracellular calcium influx [19,27,67,68,69]. One mechanism whereby CARPs have the capacity to reduce intracellular calcium influx is the reduction of neuronal cell surface ion channels and receptors; thereby reducing excitotoxic calcium influx. To this end, CARPs have been demonstrated to reduce the cell surface expression or function of neuronal NMDA receptors [23,63,70,71,72,73], voltage gated calcium channels (CaV2.2 and CaV3.3) [68,70,74,75], a voltage gated sodium channel (NaV1.7) [76] and the sodium calcium exchanger 3 (NCX3) [70]. Given their effects on different receptors, and the positive charge of CARPs, they may also be antagonizing ion channel receptor function directly and/or by affecting calcium or sodium ion influx via an electrostatic mechanism.
In addition to neuronal cell surface receptor interactions, CARPs may also exert pleiotropic neuroprotective effects by targeting mitochondria and maintaining mitochondrial integrity by reducing calcium influx into the organelle [65,77,78,79,80,81,82,83]. CARPs have the capacity to inhibit proteolytic enzymes including the proteasome [84,85], as well as pro-protein convertases [86,87,88] that activate MMPs [89]. Inhibition of the proteasome is known to be beneficial following cerebral ischemia [90,91,92]; a mechanism that could be mediated by increased activity of hypoxia-inducible factor 1-α (HIF1α) and decreased activity of nuclear factor κ-light-chain-enhancer of B cells (NFκB). Similarly, MMP activation in the brain is known to be detrimental following cerebral ischemia and inhibiting MMP activation is neuroprotective (see review Cunningham et al. [93]).
Furthermore, there is also evidence that CARPs activate pro-survival signaling pathways [67,94], modulate immune responses [95,96,97] and act as anti-oxidant molecules in their own right due to their guanidine group in arginine residues [65,83,98,99,100,101].
Interestingly, the neuroprotective potency of CARPs appears to correlate with the same characteristic that improve the ability of cationic CPPs to target and transverse cellular membranes [19,29]. This suggests that CARP-mediated neuroprotection is associated with cellular membrane interactions (e.g., plasma and mitochondrial), anionic structures (phospholipids/cardiolipin, heparan sulfate proteoglycans, chondroitin sulfate proteoglycans, sialic acid resides, anionic regions of surface receptors) and/or increased levels of cellular uptake, which enable the peptides to interact with membrane (e.g., ion channel receptors) and intracellular targets (e.g., proteins, ROS and mitochondria).

4.4. Studies Using CARPs in Animal Models of HIE

All descriptive details of the studies discussed below are presented in chronological order of publication (Table 1).

4.4.1. TAT-NEMO Binding Domain (NBD)

NEMO binding domain (NBD) is an 11-amino acid peptide (NBD: TALDWSWLQTE735–745; net charge −2) derived from the Nuclear Factor-κβ (NF-κβ) Essential Modulator (NEMO)/IKK-γ-Binding Domain. The NBD peptide was designed to inhibit the activation of NFκB, a transcription factor involved in many stress responses. For example, NBD was originally developed to suppress inflammatory responses (e.g., IL-6, iNOS, MMP-9 and COX-2) and the regulation of apoptotic Bcl proteins (e.g., Bcl-2 and Bcl-x) following cytokine stimulation in several disorders including inflammatory bowel disease, arthritis, type I diabetes, multiple sclerosis and Parkinson’s disease [102]. Activation of NF-κB requires upstream kinase complex formation (IκB-kinase; IKK) composed of the IKKα, IKKβ and NEMO proteins. For NF-κB activation, NEMO interacts with a carboxyl terminal sequence within both IKK-α and IKK-β, known as the NBD. The NBD peptide inhibits NF-κB activation by blocking NEMO/IKK complex formation [103].
The first study examining the neuroprotective efficacy of the NBD peptide in a model of HIE (P12 rat) administered the peptide [(14,820 nmol/kg: intraperitoneally (IP)] immediately, 6 and 12 h after hypoxia and did not demonstrate any neuroprotection; instead NBD appeared to increase brain injury [104]. To improve delivery to the brain, the NBD peptide was fused to TAT (TAT-NBD: YGRKKRRQRRR-TALDWSWLQTE; net charge +6) and assessed in a P7 rat model of HIE [105], where administration (6917 nmol/kg: IP) immediately and 3 h, or as a single injection at 0, 3 or 6 h, after hypoxia was neuroprotective up to 6 weeks post-HI. When TAT-NBD was administered immediately and 3 h after hypoxia it prevented up-regulation of p53 and activation of caspase-3 24 h post-HI. TAT-NBD treatment also prevented the activation of IKK in the brain 3 h post-HI. Despite TAT-NBD inhibiting IKK and NF-κB activation, it had no effect on HI-induced increases in cytokines (e.g., IL-1β, TNF-α, IL-4, IL-10 and IL-1-RA). The study also assessed a TAT-NBD control peptide, where two tryptophan residues were substituted for two alanine residues (YGRKKKRRQRRR-TALDASALQTE; net charge +6) and was shown not to be neuroprotective. Further assessment of TAT-NBD (6817 nmol/kg: IP) involving administration immediately and 3 h, immediately, 3, 6 and 12 h, immediately, 6 and 12 h, as well as 18 and 21 h after hypoxia [106], revealed long-term neuroprotection only in animals treated immediately and 3 h post-HI. Concomitantly, TAT-NBD treatment immediately and 3 h after hypoxia significantly reduced HI-mediated cytokine levels, whereas immediate, 3, 6 and 12-h treatment did not reduce cytokines. In a subsequent study, TAT-NBD treatment (6818 nmol/kg: IP) immediately and 3 h after hypoxia demonstrated significant improvements in behavioral outcomes up to 14 weeks post-HI [107].
Since intrauterine infections can be associated with HIE, TAT-NBD was assessed in a lipopolysaccharide (LPS)-sensitized (IP injection) HIE model in the P7 rat [108]. Animals were administered with TAT-NBD (477 nmol/kg) intranasally (IN) 10 min after hypoxia, using three HI injury conditions; HI only, 4-h LPS-pre-treatment + HI or 72-h LPS-pre-treatment + HI. Treatment with TAT-NBD significantly reduced brain injury only in the LPS-sensitized HI models, when assessed 7 days post-HI. While it was surprising that TAT-NBD did not reduce brain injury following HI only, its neuroprotective actions in LPS-sensitized HI models may be due to the ability of the cationic TAT-NBD peptide to electrostatically bind and neutralize anionic LPS molecules or by reducing the effects of inflammatory cytokines. Neutralizing LPS would have the effect of suppressing the molecules capacity to evoke an immune response and inflammatory response. In support of this mechanism, previous studies using in vitro cell and in vivo tissue injury models have demonstrated that CARPs (e.g., HBD-2, HNP-1 and indolicidin) can bind to LPS and reduce its inflammatory effects [109]. Furthermore, it is possible that the IN delivery of TAT-NBD was enough to reduce the detrimental effects of LPS, but not ischemic injury associated with HI.

4.4.2. TAT-mGluR1

mGluR1 is a 14-amino acid peptide (mGluR1: VIKPLTKSYQGSGK929–942; net charge +3), corresponding to an intracellular region of the metabotropic glutamate receptor-1α (mGluR1α) protein that is cleaved by calpain in neurons following excitotoxicity [110]. It is proposed that TAT-mGluR1 (YGRKKRRQRRR-VIKPLTKSYQGSGK, net charge +11) blocks the ability of calpain to cleave mGluR1α and thus maintain the receptors ability to stimulate the neuroprotective phosphoinositide 3-kinase/protein kinase B (PI3K-Akt) signaling pathway and protect neurons from the toxic effects of excitotoxic calcium influx [110]. In a neuronal model of kainic acid mediated excitotoxicity, TAT-mGluR1 prevented mGluR1α truncation and reduced neuronal cell death [110]. In a P7 rat model of HIE [111] TAT-mGluR1 (58,590 nmol/kg; IP) administered 1 h before hypoxia significantly reduced cerebral infarction at 24 h post-HI.

4.4.3. c-Jun N-Terminal Kinase (JNK) Inhibitors

JNKI-1 is a 20-amino acid peptide (also referred to as JBD: RPKRPTTLNLFPQVPRSQDT157–176; net charge +3), derived from the signaling adaptor protein c-Jun N-terminal kinase protein-1 (JNKI-1) [112]. JNKs are a member of the mitogen-activated protein kinases (MAPK) family, encoded by the Jnk1, Jnk2 and Jnk3 genes, with high expression levels in neurons, especially after pathological injury (e.g., excitotoxicity, stroke, epilepsy and HI) [113]. The JNKI-1 peptide can inhibit JNK interaction with JNK-interacting protein-1 (JIP-1), blocking JNK phosphorylation and activation, thereby inhibiting downstream pro-death cellular signaling pathways [112]. JNK has emerged as a central mediator of excitotoxic damage in the developing [114,115] and developed CNS [116,117]. The JNKI-1 peptide derivatives bound to TAT, such as TAT-JNKI-1L (YGRKKRRQRRR-PP-RPKRPTTLNLFPQVPRSQDT-NH2, net charge +12) and its retro inverso D-enantiomer JNKI-1-TATD (tdqsrpvqpflnlttprkpr-pp-rrrqrrkkrgy-NH2; net charge +12, lower case indicates D-isoform amino acids) have demonstrated in vitro and/or in vivo neuroprotective properties. Studies using the JNKI-1 peptide alone (D-JNKI-1) or when fused to TAT (TAT-JNKI-1L and JNKI-1-TATD) have been assessed in neonatal HIE models.
The initial study examining the efficacy of D-JNKI-1 (tdqsrpvqpflnlttprkpr-NH2; net charge +4) in a P7 rat model of HIE when administered (76 nmol/kg: IP) 30 min before and 3, 5, 8, 12 and 20 h after hypoxia did not reveal any reduction in cerebral infarction at 24 h [115], although there was evidence for reduced calpain, caspase-3 and autophagic activation.
A subsequent study demonstrated that the TAT-fused peptide JNKI-1-TATL administered (2446 nmol/kg; IP) immediately and 3 h or 3 h after hypoxia, significantly reduced cerebral infarction at 48 h, while administration 6 h after hypoxia was ineffective [118]. When administered immediately and 3 h after hypoxia, functional benefits were observed 14 weeks post-HI. Despite improvements in cerebral infarct and functional outcomes, JNKI-1-TATL failed to prevent caspase-8-mediated cleavage of Bid, which was unexpected, as activated JNK is known to induce caspase-8 cleavage of Bid and promote mitochondrial pro-apoptotic cell death pathways; this suggests JNKI-1-TATL-mediated neuroprotection was occurring via mechanism independent of JNK suppression.
In a second study, the D-isoform peptide JNKI-1-TATD (2616 nmol/kg: IP) significantly reduced cerebral infarct volume when administered immediately, 3 or 6 h after hypoxia, but not when administered immediately and 3 h after hypoxia [119]. JNKI-1-TATD treatment also provided long-term functional improvements. It was also demonstrated that treatment with JNKI-1-TATD reduced mitochondrial levels of phosphorylated JNK, preserved mitochondrial integrity, and up-regulated anti-apoptotic proteins 24 h post-HI. The study also assessed a mitochondrial JNK scaffold inhibiting peptide, SabKIM1 (gfeslsvpspldlsgprvva-pp-rrrqrrkkrg; net charge +7) and a scrambled control (lpsvfgdvgapsrlpevsls-pp-rrrqrrkkrg; net charge +7); Sab (SH3 domain-binding protein 5) is a mitochondrial scaffold protein required for JNK kinase activity. Administration of SabKIM1 (2700 and 5555 nmol/kg: IP) immediately after hypoxia was neuroprotective, whereas the scrambled peptide (2700 nmol/kg: IP) was ineffective.
In our laboratory, administration of JNKI-1-TATD (1000 nmol/kg; IP) immediately after hypoxia resulted in a positive trend in reduced total infarct volume (15% reduction) although it did not improve behavioral outcomes 48 h post-HI [22]. It was also demonstrated that in cultured cortical neurons JNKI-1-TATD dose-dependently reduced glutamic acid mediated excitotoxic intracellular calcium influx.
While it was surprising that the SabKIM1 scrambled control peptide did not display any neuroprotection, it is possible that the amino acid sequence of the cargo peptide may have influenced neuroprotective potency. Importantly, we and others have confirmed that TAT-fused scrambled peptides such as TAT-p53DMs and TAT-NR2cts [29,66] still retain neuroprotective properties, again pointing to an active role of the TAT carrier and peptide arginine content mediated neuroprotective action.

4.4.4. TAT-BH4

BH4 is a 25-amino acid peptide derived from the Bcl-x(L) protein BH4 homology domain region (RTGYDNREIVMKYIHYKLSQRGYEW6–30, net charge +2.1). The BH4 domain is required by Bcl-x(L) to block cytochrome-c release from mitochondria and prevent MPTP opening [120], as well as inhibiting endoplasmic reticulum (ER) calcium release into the cytosol by binding to a regulatory domain of the inositol triphosphate-3 (IP3) receptor [121]; highlighting potential mechanisms whereby the BH4 peptide could exert a neuroprotective action after HI.
The TAT-BH4 (YGRKKRRQRRR-RTGYDNREIVMKYIHYKLSQRGYEW, net charge +10.1) peptide was demonstrated to reduce cerebral tissue damage and caspase-3 activation when administered intracerebroventricularlly (ICV; 5 µL/20 ng) 30 min before hypoxia in a P7 rat model of HI [122]. Interestingly, exposure of cultured adult neural stem cells to TAT-BH4 promoted axonal remodeling, suggesting the peptide exerts other effects that may be beneficial in improving recovery following HI.

4.4.5. Osteopontin (OPN) and TAT-Fused OPN Peptide (TAT-OPN)

Osteopontin (OPN) is a secreted acidic glycoprotein containing an arginine-glycine-aspartate (RGD) motif and possesses multiple cellular functions including chemotactic and cytokine-like actions. The OPN protein is expressed in various tissues including bone, placenta, pancreas and the CNS [123]. Following cerebral ischemia, OPN deficient mice demonstrate increased neurodegeneration [124] and when administered ICV, the protein is neuroprotective in several adult rodent cerebral ischemia and hemorrhagic stroke models [125,126,127,128].
Mechanisms thought to be associated with OPN-mediated neuroprotection include binding to the cell surface αvβ3 integrin receptor to stimulate pro-survival signaling (i.e., PI3K/Akt pathway), anti-inflammatory effects [126,129,130,131], inhibition of inducible NOS [132,133] STAT1 ubiquitination/degradation and by reducing MMP-9 and NF-κβ activation. Despite OPN’s demonstrated neuroprotective effects, in one study OPN deficient mice demonstrated no change in susceptibility to ischemic brain injury [126].
The first study to examine the neuroprotective efficacy of OPN in HIE used full length recombinant OPN protein (rOPN) in a P7 rat model of HI [134]. Animals treated with rOPN (0.03 or 0.1 µg per animal: ICV) 60 min after hypoxia, displayed a reduction in infarct volume at 48 h and significant improvements in behavioral outcomes at 7 weeks post-HI. In contrast, treatment with rOPN (0.05 µg/animal: ICV or 1.2 µg/animal: IN) or thrombin cleaved rOPN (1.2 µg/animal: IN) was not neuroprotective in a P5 mouse model of HI [135]. To improve cerebral delivery of OPN, a shorter peptide containing the RGD motif (IVPTVDVPNGRGDSLAYGLR134–153; net charge 0) was developed. However, the shorter peptide was not neuroprotective when administered ICV (0.2 µg/animal) or IN (30 µg/animal) before hypoxia in P5 mice [135]. In a subsequent study the OPN134–153 peptide was fused to TAT (TAT-OPN; YGRKKRRQRRR-IVPTVDVPNGRGDSLAYGLR, net charge +8) and assessed in a P9 mouse model of HI [136] using several peptide treatment regimens consisting of multiple dosing using IN, IP or ICV administration (see Table 1 for details). No treatment regimen reduced infarct volume or improved functional assessments up to 35 days post-HI. Taken together, current evidence indicates that at the dosing schedules examined for the TAT-OPN peptide, it is not neuroprotective in HIE rodent models.

4.4.6. P5-TAT

P5 is a 24-amino acid peptide derived from the C-terminal region of the p35 protein (KEAFWDRCLSVINLMSSKMLQINA254–277, net charge +0.9). Following cleavage of p35 by calpain, a cleavage fragment known as p25 can bind to cyclin-dependent kinase 5 (Cdk5) and alter its cellular location and stimulate its activity [137], which is known to be associated with neuronal cell death [138,139,140]. Administration (3,660, 7,320 and 9,761 nmol/kg: IP) of a modified P5 peptide (TFP5: KEAFWDRCLSVINLMSSKMLQINAYARAARRAARR; net charge +5.9) can improve outcomes in Parkinson’s disease, Alzheimer’s disease, and adult ischemic stroke models in rodents [140,141,142].
A dose-response assessment (0.23, 2.30, 5.76 and 11.5 nmol/kg; IP) of P5-TAT (KEAFWDRCLSVINLMSSKMLQINA-YGRKKRRQRRR, net charge +8.9) when administered 30 min before hypoxia in P7 rats demonstrated that all doses, except 0.23 nmol/kg, significantly reduced cerebral infarct volume at 48 h [143]. In addition, P5-TAT, when administered (11.5 nmol/kg; IP) 24 h after hypoxia, significantly improved behavioral outcomes up to 3 weeks post-HI. However, P5-TAT treatment following HI did not have any effect on the levels of Cdk5 activators, p35, p25 or p29, despite suppressing cleavage of caspase-3.

4.4.7. D-TAT-GESV

GESV is a 4-amino acid peptide (GESV, net charge −1) derived from the carboxyterminal PDZ motif (PDZ-G(D/E)XV; net charge −1) of nNOS. Proteins containing PDZ domains play a key role in anchoring receptor proteins in the membrane to intracellular cytoskeletal structures (e.g., actin and microtubules) and cytosolic proteins. Following NMDA receptor stimulation, nitric oxide synthase-1 adaptor protein (NOS1AP) via its PDZ binding domain, binds nNOS and recruits the protein to the cell membrane where it generates nitric oxide (NO). The D-TAT-GESV peptide (ygrkkrrqrrr-GESV, net charge +7) inhibits NOS1AP/nNOS binding, and is believed to confer neuroprotection by reducing NO production and activation of the p38 MAPK stress pathway [144]. In a P7 rat model of HIE, D-TAT-GESV (100 ng: ICV), administered before hypoxia reduced cerebral infarction at 7 days post-HI [145]. In comparison, the control peptide D-TAT-GASA (100 ng: ICV) was ineffective [145], which is not surprising since alanine residues can reduce CARP neuroprotective efficacy [19].

4.4.8. TAT-NR2B9c/NA-1

The development of NR2B9c as a neuroprotective agent has recently been discussed in detail [146]. Briefly, NR2B9c is a 9 amino acid peptide (KLSSIESDV1479–1487) derived from the intracellular terminal carboxyl region of the NMDA receptor NR2B subunit protein, a site that binds the adaptor protein postsynaptic density-95 (PSD-95). PSD-95 couples nNOS to the NR2B subunit and following NMDA receptor activation stimulates NO generation. The NR2B9c peptide was developed on the basis that it blocks NR2B subunit and PSD-95 binding and thereby inhibiting NO production following NMDA receptor stimulation.
TAT-NR2B9c (YGRKKRRQRRR-KLSSIESDV; net charge +7) administered (19,850 nmol/kg; IP) either 110 min or 20 min before hypoxia in mice reduced brain injury at 24 h post-HI [147]. In addition, TAT-NR2B9c treatment significantly improved behavioral outcomes up to 7 days after HI. Interestingly, a 3000 nmol/kg dose (IP), administered 20 min before hypoxia was ineffective.

4.4.9. Poly-Arginine-18 (R18 and R18D)

R18 (RRRRRRRRRRRRRRRRRR; net charge +18) and its D-enantiomer (rrrrrrrrrrrrrrrrrr; net charge +18) is an 18-amino acid peptide and has been demonstrated to have potent neuroprotective properties in various in vitro and in vivo stroke related injury models [18,24,26,29]. Moreover, we have recently demonstrated for the first time that R18 and R18D (30, 100, 300 and 1000 nmol/kg; IP), administered immediately after hypoxia, reduced cerebral infarct volume and improved behavioral outcomes at 48 h post-HI [22]. In an additional study, administration of R18D 30 (10, 30, 300 and 1000 nmol/kg; IP) and 60 min (30 nmol/kg; IP) after hypoxia, reduced cerebral infarct volume and improved behavioral outcomes at 48 h post-HI [21].

4.5. Other Peptides Examined in Animal Models of HIE

Several other peptides, not fused to a CPP, have also been examined in animal models of HIE. All descriptive details of the studies discussed below are presented in chronological order of publication and in Table 1.

4.5.1. COG133

COG133 is a 20-amino acid peptide (TEELRVRLASHLRKLRKRLL133–152, net charge +5.1) derived from the apolipoprotein E (apoE) protein. COG133 has been demonstrated to be neuroprotective in several acute brain injury models and demonstrated to possess anti-oxidant, anti-excitotoxic and anti-inflammatory properties [148,149,150,151,152]. Exactly how the COG133 peptide imparts its anti-oxidant, anti-excitotoxic and anti-inflammatory properties is not known, but the positive charge of the peptide has been considered as a critical factor [19,148]. When examined in a P7 rat model of HIE, COG133 administration (40, 200, 300, 400 and 2000 nmol/kg: ICV) immediately before hypoxia displayed a neuroprotective dose-response effect [151].

4.5.2. Connexin 43 (Cx43) Derived Peptides

Astrocytic gap junction hemichannels play an essential role in the regulation of electrolytes and metabolites between the cytosol and extracellular space. Under physiological conditions, hemichannels are usually in a closed state. Specifically, connexin 43 (Cx43) hemichannel opening occurs following hypoxia, ischemia and OGD [153,154,155,156], resulting in a disruption to cellular homeostasis and an exacerbation of excitotoxicity [157]. The non-specific blocking of gap junction hemichannels with octanol and carbenoxolone demonstrates neuroprotective effects in in vitro models of hippocampal hypoxic-hypoglycemic injury [158,159]. To overcome the non-specific blocking of gap junction hemichannels, small mimetic peptides that bind to the extracellular loop of Cx43 hemichannels, blocking hemichannel opening have been developed [160].
Several studies have assessed the administration of a Cx43 mimetic peptide (Peptide 5: VDKFLSRPTEKT, net charge +1) in fetal sheep models of HIE. Administration of Peptide 5 continuously for either 1 (50,000 nmol/kg/h; ICV) or 25 h (50,000 nmol/kg over 1 h and then over 24 h; ICV), beginning 90 min after hypoxia, improved electroencephalographic (EEG) recovery, reduced seizure activity and improved the return of sleep state cycling. In addition, the 25-h infusion of Peptide 5 improved oligodendrocyte survival at 7 days post-HI [161]. Another study demonstrated Peptide 5 administration immediately after hypoxia (50,000 nmol/kg over 1 h and then over 24 h; ICV) improved neuronal and oligodendrocyte cell survival, whereas peptide administration beginning during hypoxia (50,000 nmol/kg for 1 h and then for 24 h; ICV) was not neuroprotective [162]. In contrast, a continuous high dose infusion of Peptide 5 (50,000 nmol/kg/h over 25 h: ICV) immediately after hypoxia did not improve neurological outcome and contributed to cerebral edema in the fetal sheep 7 days post-HI [163]. In a pre-term (103 days gestation) fetal sheep model of HI, Peptide 5 administration (50,000 nmol/kg for 1 h and then for 24 h; ICV) commencing 90 min after hypoxia improved EEG recovery, reduced neuronal death and increased oligodendrocyte cell survival 7 days post-HI [164]. A study using term fetal sheep assessed Peptide 5 and hypothermia (32 ± 1 °C; 72 h), as individual treatments and when combined. Peptide 5 administration 3 h after hypoxia (50,000 nmol/kg for 1 h and then for 24 h; ICV) and hypothermia treatments alone, improved EEG power, neuronal and oligodendrocyte cell survival at 7 days post-HI. However, combined Peptide 5 and hypothermia treatment did not result in any additional improvement in EEG power or histological outcomes [165]. This suggests that neuroprotective targets, other than hemichannels, may be more appropriate to improve the therapeutic effects of hypothermia for HIE.
Other Cx43 mimetic peptides (Gap 26: VCYDKSFPISHVR; net charge +1 and Gap 27: SRPTEKTIFII; net charge +1) have demonstrated neuroprotective efficacy in a P7 rat model of HIE [166]. Gap 26 (0.64, 3.22, 6.44, 16.1 and 32.2 nmol/kg; IP) and Gap 27 (16.1 nmol/kg; IP) administered 1 h before hypoxia, reduced infarct volume at 48 h post-HI; however, the two lower doses of Gap 26 were ineffective. In addition, multiple administrations of Gap 26 (32.2 nmol/kg: IP) every day for 7 days, beginning 24 h after hypoxia reduced brain injury and improved neurological function, up to 3 weeks post-HI.
Since Cx43 mimetic peptides Peptide 5, Gap26 and Gap 27 have only one arginine residue and low cationic charge, it would seem unlikely the peptides are operating via a CARP mechanism of action.

4.5.3. Apelin-36

Apelin is an endogenous ligand of the apelin receptor, a G protein-coupled receptor, which is widely expressed throughout the body, including neurons and oligodendrocytes [167]. The human APELIN gene encodes a 77 amino acid pre-protein, that is cleaved into at least three biologically active fragments, Apelin-13(65–77), Apelin-17(61–77) and Apelin-36(42–77). The active apelin fragments are widely expressed in the body including the CNS [168]. Apelin peptides have been demonstrated to reduce neuronal excitotoxicity and activate extracellular signal-regulated kinase 1/2 ERK-1/2 and PI3K/Akt cell survival pathways in cultured neurons [169,170,171]. Apelin-36 (LVQPRGSRNGPGPWQGGRRKFRRQRPRLSHKGPMPF42–77; net charge +10.1) has been assessed in a P7 rat model of HIE [172] with administration (1000 ng: IP) 1 h before hypoxia reduced infarct volume at 48 h post-HI, and improving neurological function up to 14 days post-HI.

4.5.4. Innate Defense Regulator (IDR) Peptide IDR-1018

Innate defense regulator (IDR) peptides are endogenous cationic host defense peptides [173,174]. IDRs modulate inflammatory responses by increasing the recruitment of immune cells to sites of infection and inflammation; increasing wound-healing properties. It is believed that IDRs reduce cerebral injury following HI by reducing the impact of damaging inflammatory responses. IDR-1018 (VRLIVAVRIWRR-NH2; net charge +5) is a synthetic host defense peptide examined in an LPS-sensitized model of HIE in P9 mice [175]. IDR-1018 administered (5208 nmol/kg: IP) 4 h before hypoxia reduced brain injury-related gene pathways that include p53 activation and calcium signaling. In addition, IDR-1018 administered (5208 nmol/kg: IP) 3 h after hypoxia, reduced infarct and inflammatory mediators (e.g., TNF-α, IL-1β, IL-4, IL-6, IL-10, IL-17, keratinocyte chemoattractant, INF-γ and granulocyte-macrophage colony stimulating factor) 8 days post-HI.

5. Do All CARPs Including TAT-Fused Peptides Share a Common Neuroprotective Mechanism of Action?

Based on previous studies in our and other laboratories, examining the neuroprotective properties of various CARPs, we view it highly likely that the neuroprotection demonstrated by the CARPs described above (i.e., TAT-NBD, TAT-mGluR1, JNKI-1-TATL, JNKI-1-TATD, SabKIM1, TAT-BH4, P5-TAT, D-TAT-GESV, TAT-NR2B9c, R18, R18D, COG133, Apelin-36 and IDR-1018) is largely mediated by peptide arginine content and positive charge [19,27,29,65,66]. There are several lines of evidence that support this hypothesis, which we provide below. In addition, we previously provided an explanation why we believe a CARP-mediated neuroprotective mechanism of action is operating for the TAT-NR2B9c and JNKI-1-TATD peptides, rather than the mechanism of action in which they were originally developed [27].
It has been demonstrated that CARPs inhibit neuronal excitotoxic calcium [19,22,23,25,27,28] and potassium [64,75] influx and in doing so inhibit many of the pathophysiological processes downstream from the ischemic excitotoxic cascade (e.g., calpain activation, mitochondrial dysfunction and ROS production, calcium induced cell signaling and lipase and DNase activation). Therefore, given that the vast majority of the proposed mechanisms of action of developed TAT-fused and other neuroprotective CARPs are molecular targets downstream from excitotoxicity (e.g., NF-κB, JNK and Cdk5), it is not surprising that they are effective and appear to be inhibiting their intended targets.
Interestingly, in addition to the inhibition of excitotoxic cationic ion influx, CARPs also have the capacity to modulate other cellular processes that are neuroprotective following HI such as proteolytic enzyme activation (e.g., proteasome and MMPs), ERK-1/2 and Akt phosphorylation and the modulation of immune responses. For example, in the case of TAT-NBD, which was developed to block NF-κB activation, the ability of CARPs to inhibit the proteasome [84,85,176,177] would have the effect of preventing NF-κB activation by preventing proteasomal degradation of the NF-κB inhibitor protein i-κB.
In the case of Apelin-36, other CARPs, such as protamine and R9 (ALX40-4C) can also bind to the apelin receptor [178,179]. Therefore, it is possible that CARPs, in addition to Apelin-36, may also be able to stimulate pro-cell survival ERK-1/2 and Akt pathways [180]. Although it should be mentioned that not surprisingly, Apelin-36 can also reduce excitotoxic calcium influx [169,170,171]. In the case of CARP-mediated immune modulation, mR18L (Ac-GFRRFLGSWARIYRAFVG-NH2; net charge +4) is known to reduce LPS-induced systemic infection [181], dRK6 (RRKRRR; net charge +6) inhibits TNFα and IL-6 production by monocytes [182] and hBD3-3 (GKCSTRGRKCCRRKK; net charge +8) downregulates NF-κB induced inflammation [183]. Thus, CARPs have the potential to suppress many of the toxic inflammatory responses associated with HIE.
Future studies examining the efficacy of putative neuroprotective peptides fused to TAT would benefit with respect to confirming neuroprotective mechanisms of action by fusing the peptide to a non-cationic CPP (e.g., MTS; AAVALLPAVLLALLP) and/or replacing any negatively charged aspartate (D) and glutamate (E) residues within the peptide sequence with arginine residues; we predict these two manipulations will decrease and increase neuroprotective efficacy, respectively. It is also advisable to confirm if the TAT-fused peptide has the capacity to reduce excitotoxic calcium influx.

6. Conclusions

There is now a large body of data from various sources demonstrating that CARPs comprise a class of peptide with significant neuroprotective potential for development as an adjuvant to hypothermia, or when hypothermia cannot be applied, for the treatment of HIE. While further studies are required to elucidate neuroprotective mechanisms, available evidence indicates that CARP neuroprotection is determined by the peptide’s arginine content and cationic charge and more specifically by the guanidine head group in the arginine residues. Interestingly, CARP neuroprotective potency appears to correlate with the same characteristics that improves the ability of the cationic CPPs to target and traverse cellular membranes [19,29]. Taken together it is clear that CARPs possess many properties enabling them to target both intracellular and extracellular damaging and protective pathways that are likely to be beneficial following HIE, not to mention their ability to cross the BBB and enter cells. In conclusion, CARPs, including poly-arginine peptides, offer an exciting therapeutic approach for HIE with the potential to reduce the severity of brain injury via potentially several neuroprotective mechanisms of action.

7. Future Directions

There are several further considerations to be addressed before translation of CARPs as a therapeutic for HIE in the clinic. It will be important to firstly identify a CARP that has high efficacy and the greatest potential to be effective in HIE, in both pre-term and term infants. In addition, preclinical studies will need to examine CARP treatment in combination with hypothermia to ensure safety, and to identify any synergistic effects in terms of improving patient outcomes. It is also important that beneficial results obtained in rodent models of HI, especially with combining CARPs and hypothermia, are confirmed in a large animal model (e.g., piglet or lamb), allowing for assessment in a gyrencephalic brain with greater comparability to the human and that the efficacy of IV administration should be established prior to clinical trials.

Author Contributions

Conceptualization, A.B.E. & B.P.M.; Methodology, A.B.E. & B.P.M.; Writing—Original Draft Preparation, A.B.E. & B.P.M.; Writing—Review and Editing, A.B.E., R.S.A., N.W.K. & B.P.M. Visualization, A.B.E., R.S.A., N.W.K. & B.P.M.; Supervision, R.S.A, N.W.K. & B.P.M.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge the feedback on the manuscript provided by Frank Mastaglia.

Conflicts of Interest

B.P.M. and N.W.K. 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.

References

  1. Chalak, L.F.; Rollins, N.; Morriss, M.C.; Brion, L.P.; Heyne, R.; Sánchez, P.J. Perinatal acidosis and hypoxic-ischemic encephalopathy in preterm infants of 33 to 35 weeks’ gestation. J. Pediatr. 2012, 160, 388–394. [Google Scholar] [CrossRef] [PubMed]
  2. Wall, S.N.; Lee, A.C.; Carlo, W.; Goldenberg, R.; Niermeyer, S.; Darmstadt, G.L.; Keenan, W.; Bhutta, Z.A.; Perlman, J.; Lawn, J.E. Reducing intrapartum-related neonatal deaths in low- and middle-income countries-what works? Semin. Perinatol. 2010, 34, 395–407. [Google Scholar] [CrossRef] [PubMed]
  3. Chowdhury, H.R.; Thompson, S.; Ali, M.; Alam, N.; Yunus, M.; Streatfield, P.K. Causes of neonatal deaths in a rural subdistrict of Bangladesh: Implications for intervention. J. Health Popul. Nutr. 2010, 28, 375–382. [Google Scholar] [CrossRef] [PubMed]
  4. Simiyu, I.N.; Mchaile, D.N.; Katsongeri, K.; Philemon, R.N.; Msuya, S.E. Prevalence, severity and early outcomes of hypoxic ischemic encephalopathy among newborns at a tertiary hospital, in northern Tanzania. BMC Pediatr. 2017, 17, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lawn, J.; Shibuya, K.; Stein, C. No cry at birth: Global estimates of intrapartum stillbirths and intrapartum-related neonatal deaths. Bull. World Health Organ. 2005, 83, 409–417. [Google Scholar] [PubMed]
  6. Lee, A.C.; Kozuki, N.; Blencowe, H.; Vos, T.; Bahalim, A.; Darmstadt, G.L.; Niermeyer, S.; Ellis, M.; Robertson, N.J.; Cousens, S.; et al. Intrapartum-related neonatal encephalopathy incidence and impairment at regional and global levels for 2010 with trends from 1990. Pediatr. Res. 2013, 74 (Suppl. 1), 50–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Perlman, J.M. Intrapartum hypoxic-ischemic cerebral injury and subsequent cerebral palsy: Medicolegal issues. Pediatrics 1997, 99, 851–859. [Google Scholar] [CrossRef] [PubMed]
  8. Kolevzon, A.; Gross, R.; Reichenberg, A. Prenatal and Perinatal Risk Factors for Autism. Arch. Pediatr. Adolesc. Med. 2007, 161, 326–333. [Google Scholar] [CrossRef] [PubMed]
  9. O’Shea, T.M. Diagnosis, treatment, and prevention of cerebral palsy. Clin. Obstet. Gynecol. 2008, 51, 816–828. [Google Scholar] [CrossRef] [PubMed]
  10. Pisani, F.; Orsini, M.; Braibanti, S.; Copioli, C.; Sisti, L.; Turco, E.C. Development of epilepsy in newborns with moderate hypoxic-ischemic encephalopathy and neonatal seizures. Brain Dev. 2009, 31, 64–68. [Google Scholar] [CrossRef] [PubMed]
  11. De Haan, M.; Wyatt, J.S.; Roth, S.; Vargha-Khadem, F.; Gadian, D.; Mishkin, M. Brain and cognitive-behavioural development after asphyxia at term birth. Dev. Sci. 2006, 9, 350–358. [Google Scholar] [CrossRef] [PubMed]
  12. Meloni, B.; Craig, A.; Milech, N.; Hopkins, R.; Watt, P.; Knuckey, N. The neuroprotective efficacy of cell-penetrating peptides TAT, penetratin, Arg-9, and Pep-1 in glutamic acid, kainic acid, and in vitro ischemia injury models using primary cortical neuronal cultures. Cell. Mol. Neurobiol. 2014, 34, 173–181. [Google Scholar] [CrossRef] [PubMed]
  13. Vaslin, A.; Rummel, C.; Clarke, P.G.H. Unconjugated TAT carrier peptide protects against excitotoxicity. Neurotox. Res. 2009, 15, 123–126. [Google Scholar] [CrossRef] [PubMed]
  14. Craig, A.J.; Meloni, B.P.; Watt, P.; Knuckey, N.W. Attenuation of Neuronal Death by Peptide Inhibitors of AP-1 Activation in Acute and Delayed In Vitro Ischaemia (Oxygen/Glucose Deprivation) Models. Int. J. Pept. Res. Ther. 2010, 17, 1–6. [Google Scholar] [CrossRef]
  15. Meade, A.J.; Meloni, B.P.; Mastaglia, F.L.; Watt, P.M.; Knuckey, N.W. AP-1 inhibitory peptides attenuate in vitro cortical neuronal cell death induced by kainic acid. Brain Res. 2010, 1360, 8–16. [Google Scholar] [CrossRef] [PubMed]
  16. Meade, A.J.; Meloni, B.P.; Cross, J.; Bakker, A.J.; Fear, M.W.; Mastaglia, F.L.; Watt, P.M.; Knuckey, N.W. AP-1 inhibitory peptides are neuroprotective following acute glutamate excitotoxicity in primary cortical neuronal cultures. J. Neurochem. 2010, 112, 258–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Xu, W.; Zhou, M.; Baudry, M. Neuroprotection by cell permeable TAT-mGluR1 peptide in ischemia: Synergy between carrier and cargo sequences. Neuroscientist 2008, 14, 409–414. [Google Scholar] [CrossRef] [PubMed]
  18. Milani, D.; Knuckey, N.; Anderton, R.; Cross, J.; Meloni, B. The R18 Polyarginine Peptide Is More Effective Than the TAT-NR2B9c (NA-1) Peptide When Administered 60 min after Permanent Middle Cerebral Artery Occlusion in the Rat. Stroke Res. Treat. 2016, 2016, 2372710. [Google Scholar] [PubMed]
  19. Meloni, B.P.; Milani, D.; Edwards, A.B.; Amderton, R.S.; O’Hare Doig, R.L.; Fitzgerald, M.; Palmer, T.N.; Kunckey, N.W. Neuroprotective peptides fused to arginine-rich cell penetrating peptides: Neuroprotective mechanism likely mediated by peptide endocytic properties. Pharmacol. Ther. 2015, 153, 36–54. [Google Scholar] [CrossRef] [PubMed]
  20. Milani, D.; Bakeberg, M.C.; Cross, J.L.; Clark, V.W.; Anderton, R.S.; Blacker, D.J.; Knuckey, N.W.; Meloni, B.P. Comparison of neuroprotective efficacy of poly-arginine R18 and R18D (D-enantiomer) peptides following permanent middle cerebral artery occlusion in the Wistar rat and in vitro toxicity studies. PLoS ONE 2018, 13, e0193884. [Google Scholar] [CrossRef] [PubMed]
  21. Edwards, A.B.; Anderton, R.S.; Knuckey, N.W.; Meloni, B.P. Assessment of therapeutic window for poly-arginine-18D (R18D) in a P7 rat model of perinatal hypoxic-ischaemic encephalopathy. In Press. J. Neurosci. Res. 2018. [Google Scholar]
  22. Edwards, A.B.; Cross, J.L.; Anderton, R.S.; Knuckey, N.W.; Meloni, B.P. Poly-arginine R18 and R18D (D-enantiomer) peptides reduce infarct volume and improves behavioural outcomes following perinatal hypoxic-ischaemic encephalopathy in the P7 rat. Mol. Brain 2018, 11, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. MacDougall, G.; Anderton, R.S.; Edwards, A.B.; Knuckey, N.W.; Meloni, B.P. The Neuroprotective Peptide Poly-Arginine-12 (R12) Reduces Cell Surface Levels of NMDA NR2B Receptor Subunit in Cortical Neurons; Investigation into the Involvement of Endocytic Mechanisms. J. Mol. Neurosci. 2016, 61, 235–246. [Google Scholar] [CrossRef] [PubMed]
  24. Milani, D.; Clark, V.; Cross, J.; Anderton, R.; Knuckey, N.; Meloni, B. Poly-arginine peptides reduce infarct volume in a permanent middle cerebral artery rat stroke model. BMC Neurosci. 2016, 17, 19. [Google Scholar] [CrossRef] [PubMed]
  25. Chiu, L.S.; Anderton, R.S.; Cross, J.L.; Clark, V.W.; Edwards, A.B.; Knuckey, N.W.; Meloni, B.P. Assessment of R18, COG1410, and APP96-110 in excitotoxicity and traumatic brain injury. Transl. Neurosci. 2017, 8, 147–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Milani, D.; Cross, J.; Anderton, R.; Blacker, D.; Knuckey, N.; Meloni, B. Neuroprotective efficacy of poly-arginine R18 and NA-1 (TAT-NR2B9c) peptides following transient middle cerebral artery occlusion in the rat. Neurosci. Res. 2017, 114, 9–15. [Google Scholar] [CrossRef] [PubMed]
  27. Meloni, B.P.; Brookes, L.M.; Clark, V.W.; Cross, J.L.; Edwards, A.B.; Anderton, R.S.; Hopkins, R.M.; Hoffmann, K.; Knuckey, N.W. Poly-arginine and arginine-rich peptides are neuroprotective in stroke models. J. Cereb. Blood Flow Metab. 2015, 35, 993–1004. [Google Scholar] [CrossRef] [PubMed]
  28. Edwards, A.B.; Anderton, R.S.; Knuckey, N.W.; Meloni, B.P. Characterisation of neuroprotective efficacy of modified poly-arginine-9 (R9) peptides using a neuronal glutamic acid excitotoxicity model. Mol. Cell. Biochem. 2016, 426, 75–85. [Google Scholar] [CrossRef] [PubMed]
  29. Meloni, B.P.; Milani, D.; Cross, J.L.; Clark, V.W.; Edwards, A.B.; Anderton, R.S.; Blacker, D.J.; Knuckey, N.W. Assessment of the Neuroprotective Effects of Arginine-Rich Protamine Peptides, Poly-Arginine Peptides (R12-Cyclic, R22) and Arginine–Tryptophan-Containing Peptides Following In Vitro Excitotoxicity and/or Permanent Middle Cerebral Artery Occlusion in Rats. Neuromol. Med. 2017, 19, 271–285. [Google Scholar] [CrossRef] [PubMed]
  30. Novak, C.M.; Ozen, M.; Burd, I. Perinatal Brain Injury: Mechanisms, Prevention, and Outcomes. Clin. Perinatol. 2018, 45, 357–375. [Google Scholar] [CrossRef] [PubMed]
  31. Giussani, D.A. The fetal brain sparing response to hypoxia: Physiological mechanisms. J. Physiol. 2016, 594, 1215–1230. [Google Scholar] [CrossRef] [PubMed]
  32. Olney, J.W.; Price, M.T.; Samson, L.; Labruyere, J. The role of specific ions in glutamate neurotoxicity. Neurosci. Lett. 1986, 65, 65–71. [Google Scholar] [CrossRef]
  33. Choi, D.W. Excitotoxic cell death. J. Neurobiol. 1992, 23, 1261–1276. [Google Scholar] [CrossRef] [PubMed]
  34. Arai, A.; Vanderklish, P.; Kessler, M.; Lee, K.; Lynch, G. A brief period of hypoxia causes proteolysis of cytoskeletal proteins in hippocampal slices. Brain Res. 1991, 555, 276–280. [Google Scholar] [CrossRef]
  35. Zhang, J.; Dawson, V.; Dawson, T.; Snyder, S. Nitric oxide activation of poly(ADP-ribose) synthetase in neurotoxicity. Science 1994, 263, 687–689. [Google Scholar] [CrossRef] [PubMed]
  36. Stout, A.K.; Raphael, H.M.; Kanterewicz, B.I.; Klann, E.; Reynolds, I.J. Glutamate-induced neuron death requires mitochondrial calcium uptake. Nat. Neurosci. 1998, 1, 366–373. [Google Scholar] [CrossRef] [PubMed]
  37. Castilho, R.F.; Ward, M.W.; Nicholls, D.G. Oxidative stress, mitochondrial function, and acute glutamate excitotoxicity in cultured cerebellar granule cells. J. Neurochem. 1999, 72, 1394–1401. [Google Scholar] [CrossRef] [PubMed]
  38. O’Hare, M.J.; Kushwaha, N.; Zhang, Y.; Aleyasin, H.; Callaghan, S.M.; Slack, R.S.; Albert, P.R.; Vincent, I.; Park, D.S. Differential roles of nuclear and cytoplasmic cyclin-dependent kinase 5 in apoptotic and excitotoxic neuronal death. J. Neurosci. 2005, 25, 8954–8966. [Google Scholar] [CrossRef] [PubMed]
  39. Ikonomidou, C.; Kaindl, A.M. Neuronal Death and Oxidative Stress in the Developing Brain. Antioxid. Redox Signal. 2011, 14, 1535–1550. [Google Scholar] [CrossRef] [PubMed]
  40. Favié, L.M.A.; Cox, A.R.; van den Hoogen, A.; Nijboer, C.H.A.; Peeters-Scholte, C.M.P.C.D.; van Bel, F.; Egberts, T.C.G.; Rademaker, C.M.A.; Groenendaal, F. Nitric Oxide Synthase Inhibition as a Neuroprotective Strategy Following Hypoxic–Ischemic Encephalopathy: Evidence From Animal Studies. Front. Neurol. 2018, 9, 258. [Google Scholar] [CrossRef] [PubMed]
  41. Rousset, C.I.; Baburamani, A.A.; Thornton, C.; Hagberg, H. Mitochondria and perinatal brain injury. J. Matern. Fetal Neonatal Med. 2012, 25 (Suppl. 1), 35–38. [Google Scholar] [CrossRef] [PubMed]
  42. Thornton, C.; Hagberg, H. Role of mitochondria in apoptotic and necroptotic cell death in the developing brain. Clin. Chim. Acta. 2015, 451 Pt A, 35–38. [Google Scholar] [CrossRef]
  43. Lu, Y.; Tucker, D.; Dong, Y.; Zhao, N.; Zhuo, X.; Zhang, Q. Role of Mitochondria in Neonatal Hypoxic-Ischemic Brain Injury. J. Neurosci. Rehabil. 2015, 2, 1–14. [Google Scholar] [PubMed]
  44. Wang, X.; Carlsson, Y.; Basso, E.; Zhu, C.; Rousset, C.I.; Rasola, A.; Johansson, B.R.; Blomgren, K.; Mallard, C.; Bernardi, P.; et al. Developmental Shift of Cyclophilin D Contribution to Hypoxic-Ischemic Brain Injury. J. Neurosci. 2009, 29, 2588–2596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Bernardi, P.; Krauskopf, A.; Basso, E.; Petronilli, V.; Blachly-Dyson, E.; Di Lisa, F.; Forte, M.A. The mitochondrial permeability transition from in vitro artifact to disease target. FEBS J. 2006, 273, 2077–2099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Thornton, C.; Rousset, C.I.; Kichev, A.; Miyakuni, Y.; Vontell, R.; Baburamani, A.A.; Fleiss, B.; Gressens, P.; Hagberg, H. Molecular Mechanisms of Neonatal Brain Injury. Neurol. Res. Int. 2012, 2012, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Blomgren, K.; Hagberg, H. Free radicals, mitochondria, and hypoxia–ischemia in the developing brain. Free Radic. Biol. Med. 2006, 40, 388–397. [Google Scholar] [CrossRef] [PubMed]
  48. Zhu, C.; Wang, X.; Deinum, J.; Huang, Z.; Gao, J.; Modjtahedi, N.; Neagu, M.R.; Nilsson, M.; Eriksson, P.S.; Hagberg, H.; et al. Cyclophilin A participates in the nuclear translocation of apoptosis-inducing factor in neurons after cerebral hypoxia-ischemia. J. Exp. Med. 2007, 204, 1741–1748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Hagberg, H.; Mallard, C.; Ferriero, D.M.; Vannucci, S.J.; Levison, S.W.; Vexler, Z.S.; Gressens, P. The role of inflammation in perinatal brain injury. Nat. Rev. Neurol. 2015, 11, 192–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Liu, F.; McCullough, L.D. Inflammatory responses in hypoxic ischemic encephalopathy. Acta Pharmacol. Sin. 2013, 34, 1121–1130. [Google Scholar] [CrossRef] [PubMed]
  51. Algra, S.O.; Groeneveld, K.M.; Schadenberg, A.W.; Haas, F.; Evens, F.C.; Meerding, J.; Koenderman, L.; Jansen, N.J.; Prakken, B.J. Cerebral ischemia initiates an immediate innate immune response in neonates during cardiac surgery. J. Neuroinflamm. 2013, 10, 796. [Google Scholar] [CrossRef] [PubMed]
  52. Kaur, C.; Sivakumar, V.; Yip, G.W.; Ling, E.A. Expression of syndecan-2 in the amoeboid microglial cells and its involvement in inflammation in the hypoxic developing brain. Glia 2009, 57, 336–349. [Google Scholar] [CrossRef] [PubMed]
  53. Ma, H.; Sinha, B.; Pandya, R.S.; Lin, N.; Popp, A.J.; Li, J.; Tao, J.; Wang, X. Therapeutic hypothermia as a neuroprotective strategy in neonatal hypoxic-ischemic brain injury and traumatic brain injury. Curr. Mol. Med. 2012, 12, 1282–1296. [Google Scholar] [CrossRef] [PubMed]
  54. Gluckman, P.D.; Wyatt, J.S.; Azzopardi, D.; Ballard, R.; Edwards, A.D.; Ferriero, D.M.; Polin, R.A.; Robertson, C.M.; Thoresen, M.; Whitelaw, A.; et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: Multicentre randomised trial. Lancet 2005, 365, 663–670. [Google Scholar] [CrossRef]
  55. Shankaran, S.; Laptook, A.R.; Ehrenkranz, R.A.; Tyson, J.E.; McDonald, S.A.; Donovan, E.F.; Fanaroff, A.A.; Poole, W.K.; Wright, L.L.; Higgins, R.D.; et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N. Engl. J. Med. 2005, 353, 1574–1584. [Google Scholar] [CrossRef] [PubMed]
  56. Azzopardi, D.V.; Strohm, B.; Edwards, A.D.; Dyet, L.; Halliday, H.L.; Juszczak, E.; Kapellou, O.; Levene, M.; Marlow, N.; Porter, E.; et al. Moderate Hypothermia to Treat Perinatal Asphyxial Encephalopathy. N. Engl. J. Med. 2009, 361, 1349–1358. [Google Scholar] [CrossRef] [PubMed]
  57. Jacobs, S.E.; Morley, C.J.; Inder, T.E.; Stewart, M.J.; Smith, K.R.; McNamara, P.J.; Wright, I.M.; Kirpalani, H.M.; Darlow, B.A.; Doyle, L.W.; et al. Whole-Body Hypothermia for Term and Near-Term Newborns With Hypoxic-Ischemic Encephalopathy. Arch. Pediatr. Adolesc. Med. 2011, 165, 692–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Jacobs, S.E.; Berg, M.; Hunt, R.; Tarnow-Mordi, W.O.; Inder, T.E.; Davis, P.G. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst. Rev. 2013. [Google Scholar] [CrossRef] [PubMed]
  59. Hall, N.J.; Eaton, S.; Peters, M.J.; Hiorns, M.P.; Alexander, N.; Azzopardi, D.V.; Pierro, A. Mild controlled hypothermia in preterm neonates with advanced necrotizing enterocolitis. Pediatrics 2010, 125, e300–e308. [Google Scholar] [CrossRef] [PubMed]
  60. Rao, R.; Trivedi, S.; Vesoulis, Z.; Liao, S.M.; Smyser, C.D.; Mathur, A.M. Safety and Short-Term Outcomes of Therapeutic Hypothermia in Preterm Neonates 34–35 Weeks Gestational Age with Hypoxic-Ischemic Encephalopathy. J. Pediatr. 2016. [Google Scholar] [CrossRef] [PubMed]
  61. Committee on Fetus and Newborn; Papile, L.A.; Baley, J.E.; Benitz, W.; Cummings, J.; Carlo, W.A.; Eichenwald, E.; Kumar, P.; Polin, R.A.; Tan, R.C. Hypothermia and neonatal encephalopathy. Pediatrics 2014, 133, 1146–1150. [Google Scholar] [PubMed]
  62. Fosgerau, K.; Hoffmann, T. Peptide therapeutics: Current status and future directions. Drug Discov. Today 2015, 20, 122–128. [Google Scholar] [CrossRef] [PubMed]
  63. Ferrer-Montiel, A.V.; Merino, J.M.; Blondelle, S.E.; Perez-Paya, E.; Houghten, R.A.; Montal, M. Selected peptides targeted to the NMDA receptor channel protect neurons from excitotoxic death. Nat. Biotechnol. 1998, 16, 286–291. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, W.W.; Zhou, M.M.; Baudry, M. Neuroprotection by Cell Permeable TAT-mGluR1 Peptide in Ischemia: Synergy between Carrier and Cargo Sequences. Neuroscientist 2008, 14, 409–414. [Google Scholar] [CrossRef] [PubMed]
  65. Marshall, J.; Wong, K.Y.; Rupasinghe, C.N.; Tiwari, R.; Zhao, X.; Berberoglu, E.D.; Sinkler, C.; Liu, J.; Lee, I.; Parang, K.; et al. Inhibition of N-Methyl-d-aspartate-induced Retinal Neuronal Death by Polyarginine Peptides Is Linked to the Attenuation of Stress-induced Hyperpolarization of the Inner Mitochondrial Membrane Potential. J. Biol. Chem. 2015, 290, 22030–22048. [Google Scholar] [CrossRef] [PubMed]
  66. McQueen, J.; Ryan, T.J.; McKay, S.; Marwick, K.; Baxter, P.; Carpanini, S.M.; Wishart, T.M.; Gillingwater, T.H.; Manson, J.C.; Wylie, D.J.A.; et al. Pro-death NMDA receptor signaling is promoted by the GluN2B C-terminus independently of Dapk1. Elife 2017, 6, e17161. [Google Scholar] [CrossRef] [PubMed]
  67. Cook, D.R.; Gleichman, A.J.; Cross, S.A.; Doshi, S.; Ho, W.; Jordan-Sciutto, K.L.; Lynch, D.R.; Kolson, D.L. NMDA receptor modulation by the neuropeptide apelin: Implications for excitotoxic injury. J. Neurochem. 2011, 118, 1113–1123. [Google Scholar] [CrossRef] [PubMed]
  68. García-Caballero, A.; Gadotti, V.M.; Stemkowski, P.; Weiss, N.; Souza, I.A.; Hodgkinson, V.; Bladen, C.; Chen, L.; Hamid, J.; Pizzoccaro, A.; et al. The deubiquitinating enzyme USP5 modulates neuropathic and inflammatory pain by enhancing Cav3.2 channel activity. Neuron 2014, 83, 1144–1158. [Google Scholar] [CrossRef] [PubMed]
  69. Xie, J.Y.; Chew, L.A.; Yang, X.; Wang, Y.; Qu, C.; Wang, Y.; Federici, L.M.; Fitz, S.D.; Ripsch, M.S.; Due, M.R.; et al. Sustained relief of ongoing experimental neuropathic pain by a CRMP2 peptide aptamer with low abuse potential. Pain 2016, 157, 2124–2140. [Google Scholar] [CrossRef] [PubMed]
  70. Brustovetsky, T.; Pellman, J.J.; Yang, X.-F.F.; Khanna, R.; Brustovetsky, N. Collapsin response mediator protein 2 (CRMP2) interacts with N-methyl-D-aspartate (NMDA) receptor and Na+/Ca2+ exchanger and regulates their functional activity. J. Biol. Chem. 2014, 289, 7470–7482. [Google Scholar] [CrossRef] [PubMed]
  71. Sinai, L.; Duffy, S.; Roder, J.C. Src inhibition reduces NR2B surface expression and synaptic plasticity in the amygdala. Learn. Mem. 2010, 17, 364–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Tu, W.; Xu, X.; Peng, L.; Zhong, X.; Zhang, W.; Soundarapandian, M.M.; Balel, C.; Wang, M.; Jia, N.; Zhang, W.; et al. DAPK1 interaction with NMDA receptor NR2B subunits mediates brain damage in stroke. Cell 2010, 140, 222–234. [Google Scholar] [CrossRef] [PubMed]
  73. Fan, J.; Cowan, C.M.; Zhang, L.Y.J.; Hayden, M.R.; Raymond, L.A. Interaction of postsynaptic density protein-95 with NMDA receptors influences excitotoxicity in the yeast artificial chromosome mouse model of Huntington’s disease. J. Neurosci. 2009, 29, 10928–10938. [Google Scholar] [CrossRef] [PubMed]
  74. Brittain, J.M.; Chen, L.; Wilson, S.M.; Brustovetsky, T.; Gao, X.; Ashpole, N.M.; Molosh, A.I.; You, H.; Hudmon, A.; Shekhar, A.; et al. Neuroprotection against traumatic brain injury by a peptide derived from the collapsin response mediator protein 2 (CRMP2). J. Biol. Chem. 2011, 286, 37778–37792. [Google Scholar] [CrossRef] [PubMed]
  75. Feldman, P.; Khanna, R. Challenging the catechism of therapeutics for chronic neuropathic pain: Targeting CaV2.2 interactions with CRMP2 peptides. Neurosci. Lett. 2013, 557 Pt A, 27–36. [Google Scholar] [CrossRef] [Green Version]
  76. François-Moutal, L.; Dustrude, E.T.; Wang, Y.; Brustovetsky, T.; Dorame, A.; Ju, W.; Moutal, A.; Perez-Miller, S.; Brustovetsky, N.; Gokhale, V.; et al. Inhibition of the Ubc9 E2 SUMO conjugating enzyme–CRMP2 interaction decreases NaV1.7 currents and reverses experimental neuropathic pain. Pain 2018, 1. [Google Scholar] [CrossRef] [PubMed]
  77. Birk, A.V.; Chao, W.M.; Liu, S.; Soong, Y.; Szeto, H.H. Disruption of cytochrome c heme coordination is responsible for mitochondrial injury during ischemia. Biochim. Biophys. Acta 2015, 1847, 1075–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Ferré, C.A.; Davezac, N.; Thouard, A.; Peyrin, J.M.; Belenguer, P.; Miguel, M.C.; Gonzalez-Dunia, D.; Szelechowski, M. Manipulation of the N-terminal sequence of the Borna disease virus X protein improves its mitochondrial targeting and neuroprotective potential. FASEB J. 2016, 30, 1523–1533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Rigobello, M.P.; Barzon, E.; Marin, O.; Bindoli, A. Effect of polycation peptides on mitochondrial permeability transition. Biochem. Biophys. Res. Commun. 1995, 217, 144–149. [Google Scholar] [CrossRef] [PubMed]
  80. Szelechowski, M.; Betourne, A.; Monnet, Y.; Ferre, C.A.; Thouard, A.; Foret, C.; Peyrin, J.M.; Hunot, S.; Gonzalez-Dunia, D. A viral peptide that targets mitochondria protects against neuronal degeneration in models of Parkinson’s disease. Nat. Commun. 2014, 5, 5181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Szeto, H.H.; Liu, S.; Soong, Y.; Wu, D.; Darrah, S.F.; Cheng, F.Y.; Zhao, Z.; Ganger, M.; Tow, C.Y.; Seshan, S.V. Mitochondria-targeted peptide accelerates ATP recovery and reduces ischemic kidney injury. J. Am. Soc. Nephrol. 2011, 22, 1041–1052. [Google Scholar] [CrossRef] [PubMed]
  82. Zhao, K.; Zhao, G.M.; Wu, D.; Soong, Y.; Birk, A.V.; Schiller, P.W.; Szeto, H.H. Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury. J. Biol. Chem. 2004, 279, 34682–34690. [Google Scholar] [CrossRef] [PubMed]
  83. Cerrato, C.P.; Pirisinu, M.; Vlachos, E.N.; Langel, Ü. Novel cell-penetrating peptide targeting mitochondria. FASEB J. 2015, 29, 4589–4599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Anbanandam, A.; Albarado, D.C.; Tirziu, D.C.; Simons, M.; Veeraraghavan, S. Molecular basis for proline- and arginine-rich peptide inhibition of proteasome. J. Mol. Biol. 2008, 384, 219–227. [Google Scholar] [CrossRef] [PubMed]
  85. Gaczynska, M.; Osmulski, P.A.; Gao, Y.; Post, M.J.; Simons, M. Proline- and arginine-rich peptides constitute a novel class of allosteric inhibitors of proteasome activity. Biochemistry 2003, 42, 8663–8670. [Google Scholar] [CrossRef] [PubMed]
  86. Cameron, A.; Appel, J.; Houghten, R.A.; Lindberg, I. Polyarginines Are Potent Furin Inhibitors. J. Biol. Chem. 2000, 275, 36741–36749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Fugere, M.; Appel, J.; Houghten, R.A.; Lindberg, I.; Day, R. Short polybasic peptide sequences are potent inhibitors of PC5/6 and PC7: Use of positional scanning-synthetic peptide combinatorial libraries as a tool for the optimization of inhibitory sequences. Mol. Pharmacol. 2007, 71, 323–332. [Google Scholar] [CrossRef] [PubMed]
  88. Kacprzak, M.M.; Peinado, J.R.; Than, M.E.; Appel, J.; Hanrich, S.; Lipkind, G.; Houghten, R.A.; Bode, W.; Lindberg, I. Inhibition of furin by polyarginine-containing peptides: Nanomolar inhibition by nona-D-arginine. J. Biol. Chem. 2004, 279, 36788–36794. [Google Scholar] [CrossRef] [PubMed]
  89. Tian, S.; Huang, Q.; Fang, Y.; Wu, J. FurinDB: A database of 20-residue furin cleavage site motifs, substrates and their associated drugs. Int. J. Mol. Sci. 2011, 12, 1060–1065. [Google Scholar] [CrossRef] [PubMed]
  90. Doeppner, T.R.; Kaltwasser, B.; Kuckelkorn, U.; Henkelein, P.; Bretschneider, E.; Kilic, E.; Hermann, D.M. Systemic Proteasome Inhibition Induces Sustained Post-stroke Neurological Recovery and Neuroprotection via Mechanisms Involving Reversal of Peripheral Immunosuppression and Preservation of Blood-Brain-Barrier Integrity. Mol. Neurobiol. 2016, 53, 6332–6341. [Google Scholar] [CrossRef] [PubMed]
  91. Wojcik, C.; di Napoli, M. Ubiquitin-proteasome system and proteasome inhibition: New strategies in stroke therapy. Stroke 2004, 35, 1506–1518. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, W.; Hartman, R.; Ayer, R.; Maracantionio, S.; Kamper, J.; Tang, J.; Zhang, J.H. Matrix metalloproteinases inhibition provides neuroprotection against hypoxia-ischemia in the developing brain. J. Neurochem. 2009, 111, 726–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Cunningham, L.A.; Wetzel, M.; Rosenberg, G.A. Multiple roles for MMPs and TIMPs in cerebral ischemia. Glia 2005, 50, 329–339. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, Y.; Zhang, X.; Cui, H.; Zhang, C.; Zhu, C.; Li, L. Apelin-13 protects the brain against ischemia/reperfusion injury through activating PI3K/Akt and ERK1/2 signaling pathways. Neurosci. Lett. 2014, 568, 44–49. [Google Scholar] [CrossRef] [PubMed]
  95. Hilchie, A.L.; Wuerth, K.; Hancock, R.E.W. Immune modulation by multifaceted cationic host defense (antimicrobial) peptides. Nat. Chem. Biol. 2013, 9, 761–768. [Google Scholar] [CrossRef] [PubMed]
  96. Kellett, D.N. On the Anti-Inflammatory Activity of Protamine Sulphate and of Hexadimethrine Bromide, Inhibitors of Plasma Kinin Formation. Br. J. Pharmacol. Chemother. 1965, 24, 705–713. [Google Scholar] [CrossRef] [PubMed]
  97. Li, L.H.; Ju, T.C.; Hsieh, C.Y.; Dong, W.C.; Chen, W.T.; Hua, K.F.; Chen, W.J. A synthetic cationic antimicrobial peptide inhibits inflammatory response and the NLRP3 inflammasome by neutralizing LPS and ATP. PLoS ONE 2017, 12, e0182057. [Google Scholar] [CrossRef] [PubMed]
  98. Courderot-Masuyer, C.; Dalloz, F.; Maupoil, V.; Rochette, L. Antioxidant properties of aminoguanidine. Fundam. Clin. Pharmacol. 1999, 13, 535–540. [Google Scholar] [CrossRef] [PubMed]
  99. Yildiz, G.; Demiryürek, A.T.; Sahin-Erdemli, I.; Kanzik, I. Comparison of antioxidant activities of aminoguanidine, methylguanidine and guanidine by luminol-enhanced chemiluminescence. Br. J. Pharmacol. 1998, 124, 905–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Lass, A.; Suessenbacher, A.; Wölkart, G.; Mayer, B.; Brunner, F. Functional and analytical evidence for scavenging of oxygen radicals by L-arginine. Mol. Pharmacol. 2002, 61, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
  101. Mandal, S.M.; Bharti, R.; Porto, W.F.; Guari, S.S.; Mandal, M.; Franco, O.L.; Ghosh, A.K. Identification of multifunctional peptides from human milk. Peptides 2014, 56, 84–93. [Google Scholar] [CrossRef] [PubMed]
  102. Harari, O.A.; Liao, J.K. NF-κB and innate immunity in ischemic stroke. Ann. N. Y. Acad. Sci. 2010, 1207, 32–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. May, M.J.; Marienfeld, R.B.; Ghosh, S. Characterization of the Ikappa B-kinase NEMO binding domain. J. Biol. Chem. 2002, 277, 45992–46000. [Google Scholar] [CrossRef] [PubMed]
  104. Van den Tweel, E.R.; Kavelaars, A.; Lombardi, M.S.; Groenendaal, F.; May, M.; Heijnen, C.J.; van Bel, F. Selective inhibition of nuclear factor-kappaB activation after hypoxia/ischemia in neonatal rats is not neuroprotective. Pediatr. Res. 2006, 59, 232–236. [Google Scholar] [CrossRef] [PubMed]
  105. Nijboer, C.H.A.; Heijnen, C.J.; Groenendaal, F.; May, M.J.; van Bel, F.; Kavelaars, A. Strong neuroprotection by inhibition of NF-kappaB after neonatal hypoxia-ischemia involves apoptotic mechanisms but is independent of cytokines. Stroke 2008, 39, 2129–2137. [Google Scholar] [CrossRef] [PubMed]
  106. Nijboer, C.H.; Heijnen, C.J.; Groenendaal, F.; May, M.J.; van Bel, F.; Kavelaars, A. A dual role of the NF-kappaB pathway in neonatal hypoxic-ischemic brain damage. Stroke 2008, 39, 2578–2586. [Google Scholar] [CrossRef] [PubMed]
  107. Van der Kooij, M.A.; Nijboer, C.H.; Ohl, F.; Groenendaal, F.; Heijnen, C.J.; ven Bel, F.; Kavelaars, A. NF-kappaB inhibition after neonatal cerebral hypoxia-ischemia improves long-term motor and cognitive outcome in rats. Neurobiol. Dis. 2010, 38, 266–272. [Google Scholar] [CrossRef] [PubMed]
  108. Yang, D.; Syn, Y.Y.; Lin, X.; Baumann, J.M.; Dunn, R.S.; Lindquist, D.M.; Kaun, C.Y. Intranasal delivery of cell-penetrating anti-NF-κB peptides (Tat-NBD) alleviates infection-sensitized hypoxic-ischemic brain injury. Exp. Neurol. 2013, 247, 447–455. [Google Scholar] [CrossRef] [PubMed]
  109. Le Roy, D.; Di Padova, F.; Tees, R.; Lengacher, S.; Landmann, R.; Glauser, M.P.; Calandra, T.; Heumann, D. Monoclonal antibodies to murine lipopolysaccharide (LPS)-binding protein (LBP) protect mice from lethal endotoxemia by blocking either the binding of LPS to LBP or the presentation of LPS/LBP complexes to CD14. J. Immunol. 1999, 162, 7454–7460. [Google Scholar] [PubMed]
  110. Xu, W.; Wong, T.P.; Chery, N.; Gaertner, T.; Wang, Y.T.; Baudry, M. Calpain-Mediated mGluR1α Truncation: A Key Step in Excitotoxicity. Neuron 2007, 53, 399–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Zhou, M.; Xu, W.; Liao, G.; Bi, X.; Baudry, M. Neuroprotection against neonatal hypoxia/ischemia-induced cerebral cell death by prevention of calpain-mediated mGluR1alpha truncation. Exp. Neurol. 2009, 218, 75–82. [Google Scholar] [CrossRef] [PubMed]
  112. Borsello, T.; Croquelois, K.; Hornung, J.P.; Clarke, P.G. N-methyl-d-aspartate-triggered neuronal death in organotypic hippocampal cultures is endocytic, autophagic and mediated by the c-Jun N-terminal kinase pathway. Eur. J. Neurosci. 2003, 18, 473–485. [Google Scholar] [CrossRef] [PubMed]
  113. Gupta, S.; Barrett, T.; Whitmarch, A.J.; Cavanagh, J.; Sluss, H.K.; Derijard, B.; Davis, R.J. Selective interaction of JNK protein kinase isoforms with transcription factors. EMBO J. 1996, 15, 2760–2770. [Google Scholar] [PubMed]
  114. Wang, L.W.; Chang, Y.C.; Chen, S.J.; Tseng, C.H.; Tu, Y.F.; Liao, N.S.; Huang, C.C.; Ho, C.J. TNFR1-JNK signaling is the shared pathway of neuroinflammation and neurovascular damage after LPS-sensitized hypoxic-ischemic injury in the immature brain. J. Neuroinflamm. 2014, 11, 215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Ginet, V.; Puyal, J.; Magnin, G.; Clarke, P.G.H.; Truttmann, A.C. Limited role of the c-Jun N-terminal kinase pathway in a neonatal rat model of cerebral hypoxia-ischemia. J. Neurochem. 2009, 108, 552–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Yang, D.D.; Kuan, C.Y.; Whitmarch, A.J.; Rincon, M.; Zheng, T.S.; Davis, R.J.; Rakic, P.; Flavell, R.A. Absence of excitotoxicity-induced apoptosis in the hippocampus of mice lacking the Jnk3 gene. Nature 1997, 389, 865–870. [Google Scholar] [CrossRef] [PubMed]
  117. Centeno, C.; Repici, M.; Chatton, J.Y.; Riederer, B.M.; Bonny, C.; Nicod, P.; Price, M.; Clarke, P.G.; Papa, S.; Franzoso, G.; Borsello, T. Role of the JNK pathway in NMDA-mediated excitotoxicity of cortical neurons. Cell Death Differ. 2007, 14, 240–253. [Google Scholar] [CrossRef] [PubMed]
  118. Nijboer, C.H.; van der Kooij, M.A.; van Bel, F.; Ohl, F.; Heijnen, C.J.; Kavelaars, A. Inhibition of the JNK/AP-1 pathway reduces neuronal death and improves behavioral outcome after neonatal hypoxic-ischemic brain injury. Brain. Behav. Immun. 2010, 24, 812–821. [Google Scholar] [CrossRef] [PubMed]
  119. Nijboer, C.H.; Bonestroo, H.J.C.; Zijlstra, J.; Kavelaars, A.; Heijnen, C.J. Mitochondrial JNK phosphorylation as a novel therapeutic target to inhibit neuroinflammation and apoptosis after neonatal ischemic brain damage. Neurobiol. Dis. 2013, 54, 432–444. [Google Scholar] [CrossRef] [PubMed]
  120. Shimizu, S.; Konishi, A.; Kodama, T.; Tsujimoto, Y. BH4 domain of antiapoptotic Bcl-2 family members closes voltage-dependent anion channel and inhibits apoptotic mitochondrial changes and cell death. Proc. Natl. Acad. Sci. USA 2000, 97, 3100–3105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Rong, Y.P.; Bultynck, G.; Aromolaran, A.S.; Zhong, F.; Parys, J.B.; De Smedt, H.; Mignery, G.A.; Roderick, H.L.; Bootman, M.D.; Distelhorst, C.W. The BH4 domain of Bcl-2 inhibits ER calcium release and apoptosis by binding the regulatory and coupling domain of the IP3 receptor. Proc. Natl. Acad. Sci. USA 2009, 106, 14397–14402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Donnini, S.; Solito, R.; Monti, M.; Balduini, W.; Carloni, S.; Cimino, M.; Bampton, E.T.; Pinon, L.G.; Nicotera, P.; Thorpe, P.E.; Ziche, M. Prevention of ischemic brain injury by treatment with the membrane penetrating apoptosis inhibitor, TAT-BH4. Cell Cycle 2009, 8, 1271–1278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Denhardt, D.T.; Guo, X. Osteopontin: A protein with diverse functions. FASEB J. 1993, 7, 1475–1482. [Google Scholar] [CrossRef] [PubMed]
  124. Schroeter, M.; Zickler, P.; Denhardt, D.T.; Hartung, H.-P.; Jander, S. Increased thalamic neurodegeneration following ischaemic cortical stroke in osteopontin-deficient mice. Brain 2006, 129, 1426–1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Doyle, K.P.; Yang, T.; Lessov, N.S.; Ciesielski, T.M.; Stevens, S.L.; Simon, R.P.; King, J.S.; Stenzel-Poore, M.P. Nasal administration of osteopontin peptide mimetics confers neuroprotection in stroke. J. Cereb. Blood Flow Metab. 2008, 28, 1235–1248. [Google Scholar] [CrossRef] [PubMed]
  126. Meller, R.; Stevens, S.L.; Minami, M.; Cameron, J.A.; King, S.; Rosenzweif, H.; Doyle, K.; Lessov, N.S.; Simon, R.P.; Stenzel-Poore, M.P. Neuroprotection by osteopontin in stroke. J. Cereb. Blood Flow Metab. 2005, 25, 217–225. [Google Scholar] [CrossRef] [PubMed]
  127. Jin, Y.; Kim, I.Y.; Kim, I.D.; Lee, H.K.; Park, J.Y.; Han, P.L.; Kim, K.K.; Choi, H.; Lee, J.K. Biodegradable gelatin microspheres enhance the neuroprotective potency of osteopontin via quick and sustained release in the post-ischemic brain. Acta Biomater. 2014, 10, 3126–3135. [Google Scholar] [CrossRef] [PubMed]
  128. Wu, B.; Ma, Q.; Suzuki, H.; Chen, C.; Liu, W.; Tang, J.; Zhang, J. Recombinant Osteopontin Attenuates Brain Injury after Intracerebral Hemorrhage in Mice. Neurocrit. Care 2011, 14, 109–117. [Google Scholar] [CrossRef] [PubMed]
  129. Jin, Y.C.; Lee, H.; Kim, S.W.; Kim, I.D.; Lee, H.K.; Lee, Y.; Han, P.L.; Lee, J.K. Intranasal Delivery of RGD Motif-Containing Osteopontin Icosamer Confers Neuroprotection in the Postischemic Brain via αvβ3 Integrin Binding. Mol. Neurobiol. 2016, 53, 5652–5663. [Google Scholar] [CrossRef] [PubMed]
  130. Das, R.; Mahabeleshwar, G.H.; Kundu, G.C. Osteopontin Stimulates Cell Motility and Nuclear Factor κB-mediated Secretion of Urokinase Type Plasminogen Activator through Phosphatidylinositol 3-Kinase/Akt Signaling Pathways in Breast Cancer Cells. J. Biol. Chem. 2003, 278, 28593–28606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Gary, D.S.; Milhavet, O.; Camandola, S.; Mattson, M.P. Essential role for integrin linked kinase in Akt-mediated integrin survival signaling in hippocampal neurons. J. Neurochem. 2003, 84, 878–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Hwang, S.M.; Lopez, C.A.; Heck, D.E.; Gardner, C.R.; Laskin, D.L.; Laskin, J.D.; Denhardt, D.T. Osteopontin inhibits induction of nitric oxide synthase gene expression by inflammatory mediators in mouse kidney epithelial cells. J. Biol. Chem. 1994, 269, 711–715. [Google Scholar] [PubMed]
  133. Rollo, E.E.; Laskin, D.L.; Denhardt, D.T. Osteopontin inhibits nitric oxide production and cytotoxicity by activated RAW264.7 macrophages. J. Leukoc. Biol. 1996, 60, 397–404. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, W.; Ma, Q.; Suzuki, H.; Hartman, R.; Tang, J.; Zhang, J.H. Osteopontin reduced hypoxia-ischemia neonatal brain injury by suppression of apoptosis in a rat pup model. Stroke 2011, 42, 764–769. [Google Scholar] [CrossRef] [PubMed]
  135. Albertsson, A.M.; Zhang, X.; Leavenworth, J.; Bi, D.; Nair, S.; Qiao, L.; Hagberg, H.; Mallard, C.; Cantor, H.; Wang, X. The effect of osteopontin and osteopontin-derived peptides on preterm brain injury. J. Neuroinflamm. 2014, 11, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Bonestroo, H.J.C.C.; Nijboer, C.H.; van Velthoven, C.T.J.J.; van Bel, F.; Heijnen, C.J. The Neonatal Brain Is Not Protected by Osteopontin Peptide Treatment after Hypoxia-Ischemia. Dev. Neurosci. 2015, 37, 142–152. [Google Scholar] [CrossRef] [PubMed]
  137. Zheng, Y.L.; Amin, N.D.; Hu, Y.F.; Rudrabhatla, P.; Shukla, V.; Kanungo, J.; Kesavapany, S.; Grant, P.; Albers, W.; Pant, H.C. A 24-Residue Peptide (p5), Derived from p35, the Cdk5 Neuronal Activator, Specifically Inhibits Cdk5-p25 Hyperactivity and Tau Hyperphosphorylation. J. Biol. Chem. 2010, 285, 34202–34212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Su, S.C.; Tsai, L.-H. Cyclin-Dependent Kinases in Brain Development and Disease. Annu. Rev. Cell Dev. Biol. 2011, 27, 465–491. [Google Scholar] [CrossRef] [PubMed]
  139. Fischer, A.; Sananbenesi, F.; Pang, P.T.; Lu, B.; Tsai, L.-H. Opposing Roles of Transient and Prolonged Expression of p25 in Synaptic Plasticity and Hippocampus-Dependent Memory. Neuron 2005, 48, 825–838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Ji, Y.B.; Zhuang, P.P.; Ji, Z.; Wu, Y.M.; Gu, Y.; Gao, X.Y.; Pan, S.Y.; Hu, Y.F. TFP5 peptide, derived from CDK5-activating cofactor p35, provides neuroprotection in early-stage of adult ischemic stroke. Sci. Rep. 2017, 7, 40013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Shukla, V.; Zheng, Y.L.; Mishra, S.K.; Amin, N.D.; Steiner, J.; Grant, P.; Kesavapany, S.; Pant, H.C. A truncated peptide from p35, a Cdk5 activator, prevents Alzheimer’s disease phenotypes in model mice. FASEB J. 2013, 27, 174–186. [Google Scholar] [CrossRef] [PubMed]
  142. Binukumar, B.K.; Shukla, V.; Amin, N.D.; Grant, P.; Bhaskar, M.; Skuntz, S.; Steiner, J.; Pant, H.C. Peptide TFP5/TP5 derived from Cdk5 activator P35 provides neuroprotection in the MPTP model of Parkinson’s disease. Mol. Biol. Cell 2015, 26, 4478–4491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Tan, X.; Chen, Y.; Li, J.; Li, X.; Miao, Z.; Xin, N.; Zhu, J.; Ge, W.; Feng, Y.; Xu, X. The inhibition of Cdk5 activity after hypoxia/ischemia injury reduces infarct size and promotes functional recovery in neonatal rats. Neuroscience 2015, 290, 552–560. [Google Scholar] [CrossRef] [PubMed]
  144. Soriano, F.X.; Martel, M.A.; Papadia, S.; Vaslin, A.; Baxter, P.; Rickman, C.; Forder, J.; Tymianski, M.; Duncan, R.; Aarts, M.; et al. Specific targeting of pro-death NMDA receptor signals with differing reliance on the NR2B PDZ ligand. J. Neurosci. 2008, 28, 10696–10710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Li, L.L.; Ginet, V.; Liu, X.; Vergun, O.; Tuittila, M.; Mathieu, M.; Bonny, C.; Puyal, J.; Truttmann, A.C.; Courtney, M.J. The nNOS-p38MAPK Pathway Is Mediated by NOS1AP during Neuronal Death. J. Neurosci. 2013, 33, 8185–8201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Ballarin, B.; Tymianski, M. Discovery and development of NA-1 for the treatment of acute ischemic stroke. Acta Pharmacol. Sin. 2018, 39, 661–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Xu, B.; Xiao, A.J.; Chen, W.; Turlova, E.; Liu, R.; Barszcyk, A.; Sun, C.L.F.; Liu, L.; Tymianski, M.; Feng, Z.P.; et al. Neuroprotective Effects of a PSD-95 Inhibitor in Neonatal Hypoxic-Ischemic Brain Injury. Mol. Neurobiol. 2016, 53, 5962–5970. [Google Scholar] [CrossRef] [PubMed]
  148. Aono, M.; Bennett, E.R.; Kim, K.S.; Lynch, J.R.; Myers, J.; Pearlstein, R.D.; Warner, D.S.; Laskowitz, D.T. Protective effect of apolipoprotein E-mimetic peptides on N-methyl-D-aspartate excitotoxicity in primary rat neuronal-glial cell cultures. Neuroscience 2003, 116, 437–445. [Google Scholar] [CrossRef]
  149. Laskowitz, D.T.; Thekdi, A.D.; Thekdi, S.D.; Han, S.K.; Myers, J.K.; Pizzo, S.V.; Bennett, E.R. Downregulation of Microglial Activation by Apolipoprotein E and ApoE-Mimetic Peptides. Exp. Neurol. 2001, 167, 74–85. [Google Scholar] [CrossRef] [PubMed]
  150. Lynch, J.R.; Tang, W.; Wang, H.; Vitek, M.P.; Bennett, E.R.; Sullivan, P.M.; Warner, D.S.; Laskowitz, D.T. APOE Genotype and an ApoE-mimetic Peptide Modify the Systemic and Central Nervous System Inflammatory Response. J. Biol. Chem. 2003, 278, 48529–48533. [Google Scholar] [CrossRef] [PubMed]
  151. McAdoo, J.D.; Warner, D.S.; Goldberg, R.N.; Vitek, M.P.; Pearlstein, R.; Laskowitz, D.T. Intrathecal administration of a novel apoE-derived therapeutic peptide improves outcome following perinatal hypoxic-ischemic injury. Neurosci. Lett. 2005, 381, 305–308. [Google Scholar] [CrossRef] [PubMed]
  152. Misra, U.K.; Adlakha, C.L.; Gawdi, G.; McMillian, M.K.; Pizzo, S.V.; Laskowitz, D.T. Apolipoprotein E and mimetic peptide initiate a calcium-dependent signaling response in macrophages. J. Leukoc. Biol. 2001, 70, 677–683. [Google Scholar] [PubMed]
  153. Krysko, D.V.; Leybaert, L.; Vandenabeele, P.; D’Herde, K. Gap junctions and the propagation of cell survival and cell death signals. Apoptosis 2005, 10, 459–469. [Google Scholar] [CrossRef] [PubMed]
  154. Kondo, R.P.; Wang, S.-Y.; John, S.A.; Weiss, J.N.; Goldhaber, J.I. Metabolic Inhibition Activates a Non-selective Current Through Connexin Hemichannels in Isolated Ventricular Myocytes. J. Mol. Cell. Cardiol. 2000, 32, 1859–1872. [Google Scholar] [CrossRef] [PubMed]
  155. Decrock, E.; De Vuyst, E.; Vinken, M.; Van Moorhem, M.; Vranckx, K.; Wang, N.; Van Laeken, L.; De Brock, M.; D’Herde, K.; Lai, C.P.; et al. Connexin 43 hemichannels contribute to the propagation of apoptotic cell death in a rat C6 glioma cell model. Cell Death Differ. 2009, 16, 151–163. [Google Scholar] [CrossRef] [PubMed]
  156. Orellana, J.A.; Hernandez, D.E.; Ezan, P.; Velarde, V.; Bennett, M.V.; Giaume, C.; Saez, J.C. Hypoxia in high glucose followed by reoxygenation in normal glucose reduces the viability of cortical astrocytes through increased permeability of connexin 43 hemichannels. Glia 2010, 58, 329–343. [Google Scholar] [CrossRef] [PubMed]
  157. Ye, Z.-C.; Wyeth, M.S.; Baltan-Tekkok, S.; Ransom, B.R. Functional hemichannels in astrocytes: A novel mechanism of glutamate release. J. Neurosci. 2003, 23, 3588–3596. [Google Scholar] [CrossRef] [PubMed]
  158. Frantseva, M.V.; Kokarovtseva, L.; Naus, C.G.; Carlen, P.L.; MacFabe, D.; Velazquez, J.L.P. Specific gap junctions enhance the neuronal vulnerability to brain traumatic injury. J. Neurosci. 2002, 22, 644–653. [Google Scholar] [CrossRef] [PubMed]
  159. Frantseva, M.V.; Kokarovtseva, L.; Velazquez, J.L.P. Ischemia-Induced Brain Damage Depends on Specific Gap-Junctional Coupling. J. Cereb. Blood Flow Metab. 2002, 22, 453–462. [Google Scholar] [CrossRef] [PubMed]
  160. O’Carroll, S.J.; Alkadhi, M.; Nicholson, L.F.B.; Green, C.R. Connexin43 Mimetic Peptides Reduce Swelling, Astrogliosis, and Neuronal Cell Death after Spinal Cord Injury. Cell Commun. Adhes. 2008, 15, 27–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Davidson, J.O.; Green, C.R.; Nicholson, L.F.; O’Carroll, S.J.; Fraser, M.; Bennet, L.; Gunn, A.J. Connexin hemichannel blockade improves outcomes in a model of fetal ischemia. Ann. Neurol. 2012, 71, 121–132. [Google Scholar] [CrossRef] [PubMed]
  162. Davidson, J.O.; Green, C.R.; Nicholson, L.F.B.; Bennet, L.; Gunn, A.J. Connexin hemichannel blockade is neuroprotective after, but not during, global cerebral ischemia in near-term fetal sheep. Exp. Neurol. 2013, 248, 301–308. [Google Scholar] [CrossRef] [PubMed]
  163. Davidson, J.O.; Green, C.R.; Nicholson, L.F.B.; Bennet, L.; Gunn, A.J. Deleterious Effects of High Dose Connexin 43 Mimetic Peptide Infusion After Cerebral Ischaemia in Near-Term Fetal Sheep. Int. J. Mol. Sci. 2012, 13, 6303–6319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Davidson, J.O.; Drury, P.P.; Green, C.R.; Nicholson, L.F.; Bennet, L.; Gunn, A.J. Connexin hemichannel blockade is neuroprotective after asphyxia in preterm fetal sheep. PLoS ONE 2014, 9, e96558. [Google Scholar] [CrossRef] [PubMed]
  165. Davidson, J.O.; Rout, A.L.; Wassink, G.; Yuill, C.A.; Zhang, F.G.; Green, C.R.; Benent, L.; Gunn, A.J. Non-Additive Effects of Delayed Connexin Hemichannel Blockade and Hypothermia after Cerebral Ischemia in Near-Term Fetal Sheep. J. Cereb. Blood Flow Metab. 2015, 35, 2052–2061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Li, X.; Zhao, H.; Tan, X.; Kostrzewa, R.M.; Du, G.; Chen, Y.; Zhu, J.; Miao, Z.; Yu, H.; Kong, J.; et al. Inhibition of connexin43 improves functional recovery after ischemic brain injury in neonatal rats. Glia 2015, 63, 1553–1567. [Google Scholar] [CrossRef] [PubMed]
  167. Choe, W.; Albright, A.; Sulcove, J.; Jaffer, S.; Hesselgesser, J.; Lavi, E.; Crino, P.; Kolson, D.L. Functional expression of the seven-transmembrane HIV-1 co-receptor APJ in neural cells. J. Neurovirol. 2000, 6 (Suppl. 1), S61–S69. [Google Scholar] [PubMed]
  168. O’Carroll, A.-M.; Selby, T.L.; Palkovits, M.; Lolait, S.J. Distribution of mRNA encoding B78/apj, the rat homologue of the human APJ receptor, and its endogenous ligand apelin in brain and peripheral tissues. Biochim. Biophys. Acta Gene Struct. Expr. 2000, 1492, 72–80. [Google Scholar] [CrossRef]
  169. Zeng, X.J.; Yu, S.P.; Zhang, L.; Wei, L. Neuroprotective effect of the endogenous neural peptide apelin in cultured mouse cortical neurons. Exp. Cell Res. 2010, 316, 1773–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Cheng, B.; Chen, J.; Bai, B.; Xin, Q. Neuroprotection of apelin and its signaling pathway. Peptides 2012, 37, 171–173. [Google Scholar] [CrossRef] [PubMed]
  171. Ishimaru, Y.; Sumino, A.; Kajioka, D.; Shibagaki, F.; Yamamuro, A.; Yoshioka, Y.; Maeda, S. Apelin protects against NMDA-induced retinal neuronal death via an APJ receptor by activating Akt and ERK1/2, and suppressing TNF-α expression in mice. J. Pharmacol. Sci. 2017, 133, 34–41. [Google Scholar] [CrossRef] [PubMed]
  172. Gu, Q.; Zhai, L.; Feng, X.; Chen, J.; Miao, Z.; Ren, L.; Qian, X.; Yu, J.; Li, Y.; Xu, X.; et al. Apelin-36, a potent peptide, protects against ischemic brain injury by activating the PI3K/Akt pathway. Neurochem. Int. 2013, 63, 535–540. [Google Scholar] [CrossRef] [PubMed]
  173. Nijnik, A.; Madera, L.; Ma, S.; Waldbrook, M.; Elliott, M.R.; Easton, D.M.; Mayer, M.L.; Mullaly, S.C.; Kindrachuk, J.; Jenssen, H.; et al. Synthetic Cationic Peptide IDR-1002 Provides Protection against Bacterial Infections through Chemokine Induction and Enhanced Leukocyte Recruitment. J. Immunol. 2010, 184, 2539–2550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Wuerth, K.C.; Falsafi, R.; Hancock, R.E.W. Synthetic host defense peptide IDR-1002 reduces inflammation in Pseudomonas aeruginosa lung infection. PLoS ONE 2017, 12, e0187565. [Google Scholar] [CrossRef] [PubMed]
  175. Bolouri, H.; Savman, K.; Wang, W.; Thomas, A.; Maurer, N.; Dullaghan, E.; Fjell, C.D.; Ek, C.J.; Hagberg, H.; Hancock, R.E.; et al. Innate defense regulator peptide 1018 protects against perinatal brain injury. Ann. Neurol. 2014, 75, 395–410. [Google Scholar] [CrossRef] [PubMed]
  176. Bao, J.; Sato, K.; Li, M.; Gao, Y.; Abid, R.; Aird, W.; Simons, M.; Post, M.J. PR-39 and PR-11 peptides inhibit ischemia-reperfusion injury by blocking proteasome-mediated IκBα degradation. Am. J. Physiol. Circ. Physiol. 2001, 281, H2612–H2618. [Google Scholar] [CrossRef] [PubMed]
  177. Kloss, A.; Henklein, P.; Siele, D.; Schmolke, M.; Apcher, S.; Kuehn, L.; Sheppard, P.W.; Dahlmann, B. The cell-penetrating peptide octa-arginine is a potent inhibitor of proteasome activities. Eur. J. Pharm. Biopharm. 2009, 72, 219–225. [Google Scholar] [CrossRef] [PubMed]
  178. Zhou, N.; Fang, J.; Acheampong, E.; Mukhtar, M.; Pomerantz, R.J. Binding of ALX40-4C to APJ, a CNS-based receptor, inhibits its utilization as a co-receptor by HIV-1. Virology 2003, 312, 196–203. [Google Scholar] [CrossRef]
  179. le Gonidec, S.; Chaves-Almagro, C.; Bai, Y.; Kang, H.J.; Smith, A.; Wangecg, E.; Huang, X.P.; Prats, H.; Knibiehler, B.; Roth, B.L.; et al. Protamine is an antagonist of apelin receptor, and its activity is reversed by heparin. FASEB J. 2017, 31, 2507–2519. [Google Scholar] [CrossRef] [PubMed]
  180. Masri, B.; Morin, N.; Pedebernade, L.; Knibiehler, B.; Audigier, Y. The apelin receptor is coupled to Gi1 or Gi2 protein and is differentially desensitized by apelin fragments. J. Biol. Chem. 2006, 281, 18317–18326. [Google Scholar] [CrossRef] [PubMed]
  181. Sharifov, O.F.; Nayyar, G.; Ternovoy, V.V.; Mishra, V.K.; Litovsky, S.H.; Palgunachari, M.N.; Gerber, D.W.; Anantharamaiah, G.M.; Gupta, H. Cationic peptide mR18L with lipid lowering properties inhibits LPS-induced systemic and liver inflammation in rats. Biochem. Biophys. Res. Commun. 2013, 436, 705–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Yoo, S.A.; Bae, D.G.; Ryoo, J.W.; Kim, H.R.; Park, G.S.; Cho, C.S.; Chae, C.B.; Kim, W.U. Arginine-rich anti-vascular endothelial growth factor (anti-VEGF) hexapeptide inhibits collagen-induced arthritis and VEGF-stimulated productions of TNF-alpha and IL-6 by human monocytes. J. Immunol. 2005, 174, 5846–5855. [Google Scholar] [CrossRef] [PubMed]
  183. Lee, J.Y.; Suh, J.S.; Kim, J.M.; Kim, J.H.; Park, H.J.; Park, Y.J.; Chung, C.P. Identification of a cell-penetrating peptide domain from human beta-defensin 3 and characterization of its anti-inflammatory activity. Int. J. Nanomed. 2015, 10, 5423–5434. [Google Scholar]
Table 1. Studies using CARPs and other peptides examined in animal models of HIE.
Table 1. Studies using CARPs and other peptides examined in animal models of HIE.
Peptide/Protein Name; Net Charge aProposed Target bPeptide Sequence cInjury Model dRoute & Time before/after HI Agent Administered eDoseNP fStudy
TAT-NBD; +6NFκBTAT-TALDWSWLQTEP7 (W): CCAO/8% O2; 120 minIP: 0 & 3 h, or 0, 3, 6, 9 or 12 h6917 nmol/kgYes, up to 6 h[105]
IP: 0 & 3 h, or 0, 9 & 12 h, or 18 & 21 h6817 nmol/kgYes, only for 0 & 3 h[106]
IP: 0 & 3 h6818 nmol.kgYes[107]
P7 (W): CCAO/10% O2; 90 min
+ LPS 4 or 72 h before hypoxia
IN; 10 min477 nmol/kgYes, only for LPS[108]
TAT-mGluR1; +3CalpainTAT-VIKPLTKSYQGSGKP7 (SD): CCAO/8% O2; 150 minIP: −1 h58,590 nmol/kgYes[111]
D-JNKI-1; +4JIPtdqsrpvqpflnlttprkpr-NH2P7 (SD): CCAO/8% O2; 120 minIP, −0.5, 3, 5, 8, 12 and 20 h76 nmol/kgNo[115]
JNKI-1-TATL; +12TAT-PPRPKRPTTLNLFPQVPRSQDT-NH2P7 (W): CCAO/8% O2; 120 minIP: 0 and 3 h, or 3 h or 6 h2446 nmol/kgYes, except 6 h[118]
JNKI-1-TATD; +12
 
SabKIM1 +7
tdqsrpvqpflnlttprkpr-pp-tat-NH2
 
lpsvfgdvgapsrlpevsls-pp-tat
P7 (W): CCAO/8% O2; 120 minIP: 0, 3 or 6 h, or 0 & 3 h
 
IP: 0 h
2616 nmol/kg
 
2777 or 5555 nmol/kg
Yes except
0 & 3 h
Yes
[119]
JNKI-1-TATD; +12tdqsrpvqpflnlttprkpr-pp-tat-NH2P7 (SD): ECA/CCAO/8% O2; 150 minIP: 0 min1000 nmol/kgNo[22]
TAT-BH4; +2.1ApoptosisTAT-RTGYDNREIVMKYIHYKLSQRGYEWP7 (SD♂): CCAO/8% O2; 150 minICV: −0.5 h5 µL/20 ngYes[122]
T-OPN
 
OPN134–153; 0
 
OPN154–198; −4.9
αvβ3 integrin receptorThrombin cleaved OPN
 
IVPTVDVPNGRGDSLAYR
 
SKSRSFQVSDEQYPDATDEDLTSHMKSGESKESLDVIPVAQLLSM
P5 (C57B6/6J): CCAO/10% O2; 70 minIN: −70 min
 
IN or ICV: −70 min
 
ICV: −70 min
1.2 µg
 
IN: 30 µg/ICV: 0.2 µg
 
0.5 µg
No[135]
TAT-OPN; +8TAT-IVPTVDVPNGRGDSLAYGLRP9 (C57BL/6N): CCAO/10% O2; 50 minIN: 0 h or 0 & 3 h, or 0, 3 h, 1, 2 & 3 d
IP: 0 h, or 5, 7, 9, 11, 13, 15 d, or 0, 1, 3, 5, 7, 9, 11, 13, 15 d
ICV: 1 h
350 or 2100 ng
2746 nmol/kg
 
100 ng
No[136]
P5-TAT; +8.9P35KEAFWDRCLSVINLMSSKMLQINA-TATP7 (SD): CCAO/8% O2; 150 minIP: −1 h
IP: 24 h
0.23, 2.3, 5.76, or 11.5 nmol/kg
11.5 nmol/kg
Yes, except 0.23 nmol/kg[143]
D-TAT-GESV; +7NOSygrkkrrqrrr-GESVP7 (SD♂); CCAO/8% O2; 120 minICV: −120 min100 ng/animalYes[145]
TAT-NR2B9c; +7PSD-95TAT-KLSSIESDVP7 (CD1): CCAO/7.5%; 60 minIP: −110 or −20 min3000 nmol/kgYes[147]
R18; +18
R18D; +18
MultipleRRRRRRRRRRRRRRRRRR
rrrrrrrrrrrrrrrrrr
P7 (SD): ECA/CCAO/8% O2; 150 minIP: 0 h30, 100, 300 or 1000 nmol/kgYes[22]
R18D; +18rrrrrrrrrrrrrrrrrrIP: +0.5 h
IP: +1 h
IP: +1.5 h
10, 30, 100, 300 or 1000 nmol/kg
30 or 300 nmol/kg
30 or 300 nmol/kg
Yes, except 100 nmol/kg
Yes, except 300 nmol/kg
No
[21]
COG133; 5.1LDLRAc-TEELRVRLASHLRKLRKRLL-NH2P7 (W): CCAO/8% O2; 150 minICV: −0 h40, 200, 300, 400, or 2000 nmol/kgYes, except 300 nmol/kg[151]
Peptide 5; +1Cx43 astrocytic hemichannelVDKFLSRPTEKTGD128 (Romney/Suffolk sheep): bilateral tCCAO; 30 minICV: 1.5 h50,000 nmol/kg/h for 1 h ±
50,000 nmol/kg/24 h for 24 h
Yes[161]
ICV: −1 h or 0 h50,000 nmol/kg/h: 1 h + 50,000 nmol/kg/24 h for 24 hYes, except
−1 h
[162]
ICV: 0 h50,000 nmol/kg/h: 1 h + 50,000 nmol/kg/24 h for 24 h
50,000 nmol/kg/h for 25 h
Yes, except high continuous dose[163]
ICV: 3 h ± Hypothermia: 32 °C for 72 h; 3 h after tCCAO50,000 nmol/kg/h for 1 h + 50,000 nmol/kg/24 h for 24 hYes, no additive effect[165]
GD103 (Romney/Suffolk sheep): bilateral tCCAO for 25 minICV: 1.5 h50,000 nmol/kg/h for 1 h + 50,000 nmol/kg/24 h for 24 hYes[164]
Gap 26; +1
 
 
Gap 27; +1
VCYDKSFPISHVR
 
 
SRPTEKTIFII
P7 (SD): CCAO/8% O2; 150 minIP: −1 h
IP: daily for 1 to 7 d
 
IP: −1 h
0.64, 3.22, 6.44, 16.1, or 32.2 nmol/kg
32.2 nmol/kg
 
16.1 nmol/kg
Yes, except 0.64 and 3.22 nmol/kg
 
 
Yes
[166]
Apelin-36; +10.1Apelin receptorLVQPRGSRNGPGPWQGGRRKFRRQRPRLSHKGPMPFP7 (SD): CCAO8% O2; 150 minIP: −1 h240 nmol/kgYes[172]
IDR-1018; +5Immune modulationVRLIVAVRIWRR-NH2P9 (C57BL/6J): CCAO/10% O2; 20 min + LPS 14 h before hypoxiaIP: −4 h or 3 h5208 nmol/kgYes[175]
Bold data indicates peptides used in studies. a Net charge at pH 7, MUT: mutant peptide, SCR: scrambled peptide. b NFκB: nuclear factor kappa-light-chain-enhancer of activated B cells, JIP: c-Jun N-terminal kinase-interacting protein, NOS: nitric oxide synthase, PSD-95: postsynaptic density-95, LDLR: low-density lipoprotein receptor. c Peptides synthesized using d-amino acids are represented in lowercase, TAT = GRKKRRQRRR. d W: Wistar, SD: Sprague-Dawley, C57B/6: C57 black 6, CD1: cluster of differentiation 1, CCAO: common carotid artery occlusion, ECA: external carotid artery occlusion, tCCAO: transient common carotid artery occlusion, ♂ = male, GD = gestational day. e IP: intraperitoneal, IN: intranasal, ICV: intracerebroventricular, h: hours, min: minutes; negative (−) = treatment before hypoxia. f Neuroprotection and or positive effects.

Share and Cite

MDPI and ACS Style

Edwards, A.B.; Anderton, R.S.; Knuckey, N.W.; Meloni, B.P. Perinatal Hypoxic-Ischemic Encephalopathy and Neuroprotective Peptide Therapies: A Case for Cationic Arginine-Rich Peptides (CARPs). Brain Sci. 2018, 8, 147. https://doi.org/10.3390/brainsci8080147

AMA Style

Edwards AB, Anderton RS, Knuckey NW, Meloni BP. Perinatal Hypoxic-Ischemic Encephalopathy and Neuroprotective Peptide Therapies: A Case for Cationic Arginine-Rich Peptides (CARPs). Brain Sciences. 2018; 8(8):147. https://doi.org/10.3390/brainsci8080147

Chicago/Turabian Style

Edwards, Adam B., Ryan S. Anderton, Neville W. Knuckey, and Bruno P. Meloni. 2018. "Perinatal Hypoxic-Ischemic Encephalopathy and Neuroprotective Peptide Therapies: A Case for Cationic Arginine-Rich Peptides (CARPs)" Brain Sciences 8, no. 8: 147. https://doi.org/10.3390/brainsci8080147

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop