Next Article in Journal
Problems in Classifying Mild Cognitive Impairment (MCI): One or Multiple Syndromes?
Next Article in Special Issue
Integrity of Cerebellar Fastigial Nucleus Intrinsic Neurons Is Critical for the Global Ischemic Preconditioning
Previous Article in Journal
Alzheimer’s Disease Early Diagnosis Using Manifold-Based Semi-Supervised Learning
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Pathophysiology and Neuroprotective Strategies in Hypoxic-Ischemic Brain Injury and Stroke

Perron Institute for Neurological and Translational Sciences; Centre for Neuromuscular and Neurological Disorders, The University of Western Australia; Department of Neurosurgery, ,Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands 6009, Australia
Brain Sci. 2017, 7(8), 110; https://doi.org/10.3390/brainsci7080110
Submission received: 15 August 2017 / Accepted: 18 August 2017 / Published: 22 August 2017
Hypoxic-ischemic brain injury and stroke are closely related and devastating conditions that can affect individuals of all ages. Acute brain injury following cerebral ischemia as a result of stroke or hypoxia-ischemia, includes perinatal hypoxic-ischemic encephalopathy (HIE) and global cerebral ischemia (e.g., cardiac arrest/resuscitation, vasospasm, cerebral edema) together are one of the major causes of death and disability worldwide. For example, stroke alone affects an estimated 16 million people worldwide annually [1]. Similarly, HIE affects 1–3 and up to 10–26 per 1000 infants born in developed and developing countries respectively [2,3,4]. An additional dimension is that most survivors of stroke and hypoxic-ischemic brain injury are left with serious life-long physical and/or neurological disabilities.
Despite considerable research over many years, there are still no proven clinically effective pharmacological neuroprotective therapies capable of reducing the severity of brain injury following stroke or cerebral hypoxia-ischemia. As a consequence, the development of an effective pharmacological neuroprotective agent for individuals suffering a cerebral ischemic event remains an urgent unmet need. To make matters worse, there seems to be a widespread sentiment among some researchers, clinicians and pharmaceutical companies that continuing research focused on the development of neuroprotective agents will ultimately be met with failure at the clinical level. This sentiment reflects the fact that past trials on neuroprotective agents have failed to produce a clinically effective agent. While it goes without saying that investing time and effort in the development of an effective neuroprotective agent is high risk, the potential benefits in terms of improving patient outcomes are enormous; hence most impartial observers would agree that the benefits of the continuing search for neuroprotective agents far outweigh the risks.
In terms of strategies to increase the likelihood of success in developing a clinically effective neuroprotective agent there are a number of lines of investigation that could be explored. First and foremost are further experimental and clinical studies to improve our understanding of the pathophysiological processes involved in ischemic brain injury in order to identify new therapeutic targets. Importantly, given that many of the pathophysiological events associated with stroke and hypoxic-ischemic brain injury are still not fully elucidated, there is great potential for such studies to identify a new set of potential therapeutic targets leading to the development of agents with better prospects for effective translation into the clinical arena.
An alternative, but complementary approach, is to identify new compounds or improve the efficacy of existing experimental agents with neuroprotective properties. For example, our laboratory has recently highlighted the potential of a class of peptides known as cationic arginine-rich peptides (CARPs) as potent neuroprotective agents, with demonstrated efficacy in vitro and in animal models of stroke [5,6,7,8,9,10,11] and HIE [unpublished data]. Furthermore, another consideration in order to provide the best opportunity for success in terms of obtaining efficacy at the clinical level, is assessment of combination treatments or identification of compounds with multiple mechanisms of action targeting two or more neurodamaging and/or neuroprotective pathways. To this end, CARPs are known to reduce neuronal calcium influx and excitotoxic neuronal death [6,7,11], down-regulate calcium channel and TNF receptor proteins [12,13,14,15], target and assist in maintaining mitochondrial integrity [16,17,18,19], reduce the activity of the proteasome [20,21] and inhibit proprotein convertases that activate matrix metalloproteinases [22,23]. In addition, this class of peptide has the capacity to modulate immune responses [24,25,26,27] and activate pro-survival signalling pathways [28,29].
Ultimately, a better understanding of the pathophysiology of ischemic and hypoxic brain injury and the identification of novel therapeutic targets and neuroprotective compounds will be an essential pre-requisite for the development of new and effective neuroprotective therapies. Hence the aim of this Special Issue is to encourage the publication of new experimental and clinical findings to advance our understanding of pathogenic processes and to identify novel neuroprotective strategies for hypoxic-ischemic brain injury and stroke.

Conflicts of Interest

Bruno P. Meloni is a holder of several patents regarding the use of arginine-rich peptides as neuroprotective treatments.

References

  1. Feigin, V.L.; Forouzanfar, M.H.; Krishnamurthi, R.; Mensah, G.A.; Connor, M.; Bennett, D.A.; Moran, A.E.; Sacco, R.L.; Anderson, L.; Truelsen, T.; et al. Global Burden of Diseases, Injuries, and Risk Factors Study 2010 (GBD 2010) and the GBD Stroke Experts Group. Global and regional burden of stroke during 1990–2010: Findings from the Global Burden of Disease Study. Lancet 2014, 383, 245–254. [Google Scholar] [CrossRef]
  2. Pierrat, V.; Haouari, N.; Liska, A.; Thomas, D.; Subtil, D.; Truffer, P.; Groupe d’Etudes en Epidémiologie Périnatale. Prevalence, causes, and outcome at 2 years of age of newborn encephalopathy: Population based study. Arch. Dis. Child. Fetal Neonatal Ed. 2005, 90, F257–F261. [Google Scholar] [CrossRef] [PubMed]
  3. Logitharajah, P.; Rutherford, M.A.; Cowan, F.M. Hypoxic-ischemic encephalopathy in preterm infants: Antecedent factors, brain imaging, and outcome. Pediatr. Res. 2009, 66, 222–229. [Google Scholar] [CrossRef] [PubMed]
  4. Douglas-Escobar, M.; Weiss, M.D. Hypoxic-ischemic encephalopathy: A review for the clinician. JAMA Pediatr. 2015, 169, 397–403. [Google Scholar] [CrossRef] [PubMed]
  5. Meloni, B.P.; Craig, A.J.; Milech, N.; Hopkins, R.M.; Watt, P.M.; Knuckey, N.W. 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]
  6. 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. Arginine-rich peptides are neuroprotective in stroke models. J. Cereb. Blood Flow Metab. 2015, 35, 993–1004. [Google Scholar] [CrossRef] [PubMed]
  7. Meloni, B.P.; Milani, D.; Edwards, A.B.; Anderton, R.S.; O’Hare Doig, R.L.; Fitzgerald, M.; Palmer, T.N.; Knuckey, 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]
  8. Milani, D.; Clark, V.W.; Cross, J.L.; Anderton, R.S.; Knuckey, N.W.; Meloni, B.P. Poly-arginine peptides reduce infarct volume in a permanent middle cerebral artery rat stroke model. BMC Neurosci. 2016, 17, 19. [Google Scholar] [CrossRef] [PubMed]
  9. Milani, D.; Knuckey, N.W.; Cross, J.L.; Anderton, R.S.; Meloni, B.P. The R18 poly-arginine peptide is more effective than the TAT-NR2B9c (NA-1) peptide when administered 60 minutes after permanent middle cerebral artery occlusion in the rat. Stroke Res. Treat. 2016, 2016, 1–8. [Google Scholar] [CrossRef] [PubMed]
  10. Milani, D.; Cross, J.L.; Anderton, R.S.; Blacker, D.J.; Knuckey, N.W.; Meloni, B.P. Neuroprotective efficacy of R18 poly-arginine 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]
  11. Meloni, B.P.; Milani, D.; Cross, J.L.; Clark, V.W.; Edwards, A.B.; Anderton, R.S.; Blacker, D.J.; Knuckey, N.W.; Meloni, B.P. Assessment of the neuroprotective effects of arginine-rich protamine peptides, poly-arginine peptides (R12-cylic, R22) and arginine-tryptophan containing peptides following in vitro excitotoxicity and/or permanent middle cerebral artery occlusion in rats. Neuromol. Med. 2017. [Google Scholar] [CrossRef] [PubMed]
  12. 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. 2017, 61, 235–246. [Google Scholar] [CrossRef] [PubMed]
  13. Brittain, J.M.; Duarte, D.B.; Wilson, S.M.; Zhu, W.; Ballard, C.; Johnson, P.L.; Liu, N.; Xiong, W.; Ripsch, M.S.; Wang, Y.; et al. Suppression of inflammatory and neuropathic pain by uncoupling CRMP-2 from the presynaptic Ca2+ channel complex. Nat. Med. 2011, 17, 822–829. [Google Scholar] [CrossRef] [PubMed]
  14. Brustovetsky, T.; Pellman, J.J.; Yang, X.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]
  15. Fotin-Mleczek, M.; Welte, S.; Mader, O.; Duchardt, F.; Fischer, R.; Hufnagel, H.; Scheurich, P.; Brock, R. Cationic cell-penetrating peptides interfere with TNF signalling by induction of TNF receptor internalization. J. Cell Sci. 2005, 118, 3339–3351. [Google Scholar] [CrossRef] [PubMed]
  16. 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]
  17. 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]
  18. 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]
  19. Ferré, C.A.; Davezac, N.; Thouard, A.; Peyrin, J.M.; Belenguer, P.; Miquel, 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. FFASEB J. 2016, 30, 1–11. [Google Scholar] [CrossRef] [PubMed]
  20. 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]
  21. 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]
  22. 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]
  23. 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]
  24. 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]
  25. Hilchie, A.L.; Wuerth, K.; Hancock, R.E. Immune modulation by multifaceted cationic host defense (antimicrobial) peptides. Nat. Chem. Biol. 2013, 9, 761–768. [Google Scholar] [CrossRef] [PubMed]
  26. 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]
  27. 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]
  28. 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]
  29. 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]

Share and Cite

MDPI and ACS Style

Meloni, B.P. Pathophysiology and Neuroprotective Strategies in Hypoxic-Ischemic Brain Injury and Stroke. Brain Sci. 2017, 7, 110. https://doi.org/10.3390/brainsci7080110

AMA Style

Meloni BP. Pathophysiology and Neuroprotective Strategies in Hypoxic-Ischemic Brain Injury and Stroke. Brain Sciences. 2017; 7(8):110. https://doi.org/10.3390/brainsci7080110

Chicago/Turabian Style

Meloni, Bruno P. 2017. "Pathophysiology and Neuroprotective Strategies in Hypoxic-Ischemic Brain Injury and Stroke" Brain Sciences 7, no. 8: 110. https://doi.org/10.3390/brainsci7080110

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