Open access

Src Family Kinases in Intracerebral Hemorrhage

Written By

DaZhi Liu

Submitted: 11 September 2013 Published: 10 September 2014

DOI: 10.5772/58488

From the Edited Volume

Intracerebral Hemorrhage

Edited by Vikas Chaudhary

Chapter metrics overview

2,008 Chapter Downloads

View Full Metrics

1. Introduction

Intracerebral hemorrhage (ICH), which accounts for 2 million (10–15%) of about 15 million strokes worldwide each year [1], has very high mortality rates of 31% at 7 days, 59% at 1 year, 82% at 10 years, and greater than 90% at 16 years [2,3]. ICH is associated with increased intracranial pressure, hematoma, blood brain barrier (BBB) disruption, brain edema, neuron loss, motor deficits, cognitive impairment and high mortality in humans. The major challenges immediately after ICH are re-bleed, hematoma induced brain injury, brain edema and neurological deficits [4]. Potential treatments of ICH include slowing the initial bleeding during the first hours after onset; removing blood from the parenchyma or ventricles to eliminate both mechanical and chemical factors that cause brain injury; management of complications of blood in the brain; and supportive medical care and surgery for certain patients [5]. Since these treatments have great variability, there is currently no FDA approved treatment for ICH.

The time course after ICH can be divided into two stages (acute/injury and chronic/recovery) (Fig. 1). At the acute stage, glutamine, thrombin, TNF-α, VEGF and other endogenous molecules are rapidly released following ICH. These molecules team up leading to brain cell death and severe brain injury via multiple neurotoxicity pathways, including (1) Excitatory amino acid (AA) and NMDA receptor-mediated excitatory toxicity; (2) Thrombin and other mitogen-mediated mitogenic stress; (3) Vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs)-mediated changes of vascular permeability; (4) Cytokines-mediated inflammatory responses; and others (Fig. 1).

As time course transits into chronic/recovery stage post-ICH, the elevated molecules resolve gradually and in turn participate in neurogenesis via populating neural progenitor cells (NPCs) to fix the damaged brain tissue. The possible mechanisms include (1) Excitatory amino acid (AA) and NMDA receptor-mediated excitatory genesis; (2) Thrombin and other mitogen-mediated mitogenic growth; (3) Nerve growth factor (NGF), epidermal growth factor (EGF) and other growth factor-mediated neurogenesis; and others (Fig. 1).

Figure 1.

Src family kinases (SFKs) participate in mitogenic signaling pathways that play critical roles in blood-brain barrier (BBB) disruption and self-repair after intracerebral hemorrhage (ICH). The network does not apply to certain cell type(s). The arrows do not necessarily indicate direct binding and/or activation of the downstream molecules; intermediate proteins or kinases may exist.

Src family kinases (SFKs), a family of proto-oncogenic proteins, participate in both neuro-toxicity at acute stage and neurogenesis during recovery stage post-ICH (Fig 1). Our previous studies have confirmed the time-specific and conflicting roles of SFKs using their inhibitor (PP2) after ICH: (1) Acute administration of PP2 (immediately post-ICH) decreases local cerebral glucose utilization (LCGU), activity of SFKs, attenuates BBB breakdown, brain edema, and cell death around ICH and improves behavioral function following ICH [6-9]; (2) Chronic inhibition of PP2 (2-6 days) blocks BBB repair and brain edema resolution in the recovery stage (7-14 days) after ICH [9].

Advertisement

2. Tissue specificity, structure and activity regulation of SFKs

SFKs are a family of non-receptor protein tyrosine kinases, include nine family members Src, Fyn, Lck, Lyn, Yes, Hck, Blk, Fgr, and Yrk [10-12], of which Src, Fyn, Yes and Yrk widely expressed whereas the rest members are expressed in specific tissues [13]. In addition, one tissue can express multiple SFK members, for example, Src, Fyn, Yes, and Lck have been examined in brain [13-19]. Importantly, the different SFK family members often compensate for one another [20], which are supported by the evidence that the mice deficient in Src can survive though Src plays vital role in cell signaling transduction [20]. SFKs share a conserved domain structure consisting of consecutive SH3 (polyproline type II helix for protein-protein interaction), SH2 (phosphotyrosine recognition), and SH1 (tyrosine kinase catalytic activity) [12]. All SFK family members also contain an membrane-targeting region at their N-terminus that is followed by a unique domain of 50–70 residues, and the unique region is divergent among family members [12]. Although it still remains incompletely clear, Src activity is regulated by tyrosine phosphorylation at two sites (one is at Tyr416 in the SH1 domain, the other at Tyr527 in the short C-terminal tail), but with opposing effects. While phosphorylation at Tyr416 activates Src, phosphorylation at Tyr527 inactivates Src [13,21].

Advertisement

3. SFKs modulate NMDA receptor for brain injury after ICH

NMDA receptors are ionotropic glutamate receptors, comprise NR1, NR2 and NR3 subunits, which form the central conductance pathway [22,23]. In the physiological conductions, activation of NMDA receptors results in the opening of an ion channel that allows the flow of Na+ and small amounts of Ca2+ into the cell and K+ out of the cell [22,23]. Following ICH there is a transient increase of glutamate release and local cerebral glucose utilization in the region surrounding the ICH, and the antagonists of NMDA receptors reverse the glucose hyper-metabolism produced by ICH [6]. However, glutamate alone could not explain the hypermetabolism since glutamate injected directly into brain did not produce hypermetabolism [6]. Apart from glutamate release, ICH may affect NMDA receptors in some way to make them more sensitive to glutamate in order to mediate injury and/or hypermetabolism.

ICH activates SFKs [7,8], and SFK members (e.g., Src, Fyn) up-regulate the ion channel activity of NMDA receptor and make them more sensitive to glutamate by phosphorylating the NR2A and NR2B subunits of the NMDA receptors [19,24-27]. It has been proved that phosphorylation by Src at Tyr-1292, Tyr-1325 and Tyr-1387 in NR2A subunit increases activity of NMDA receptors, and phosphorylation of tyrosine residues by Src in the C-terminal of the subunits prevents a Zn2+-dependent inhibition of the NMDA receptors and thus increases channel conductivity [28-30]. We have demonstrated that either NMDA receptor inhibitors (MK-801) or SKF inhibitors (PP2) can attenuate brain injury at the acute stage after ICH (Fig. 2) [8]. These results suggest that either activation of NMDA receptors or SFKs is sufficient to produce brain injury post-ICH.

Figure 2.

The effects of graded doses of SFK inhibitor PP2 (0.3 and 1.0 mg/kg, i.p.) and NMDA receptor inhibitor MK801 (1.0 mg/kg, i.p.) on injury produced by thrombin injections into striatum of Sprague-Dawley rats. A. Representative Hematoxylin-Eosin stained section shows that 20U of thrombin causes brain injury, including hematoma and edema. B. Control injections of BSA into striatum produced minor injury. C. Infarction volumes 24h following striatum injections of thrombin compared to control BSA injections (BSA/Control) (n=6). Several groups of animals received striatal injections of thrombin: just thrombin alone (Throm/Saline) (n=6); prior intraperitoneal injection of PP2 (Throm/PP2/0.3mg/kg) (n=6); prior intraperitoneal injection of a higher dose of PP2 (Throm/PP2/1.0mg/kg) (n=6); and prior intraperitoneal injection of MK801 (Throm/MK801/1.0mg/kg) (n=6).. * p<0.05 and ** P<0.01 vs Throm/Saline (one-way ANOVA followed by Dunnett’s post hoc test). D. PP2 (0.3 and 1.0 mg/kg, i.p.) and MK801 (1.0 mg/kg, i.p.) decrease the thrombin-induced motor deficits (n=9 for each group) using Elevated Body Swing Test (EBST) 23.5 hours after thrombin injections. Biased swinging behavior was calculated as follows: L/ (L+R) (%) for left biased swings (L, left-biased swings; R, right-biased swings). * p<0.05 and ** P<0.01 vs Throm/Saline (one-way ANOVA followed by Dunnett’s post hoc test).

NMDA receptor activation has also been shown to enhance NPCs proliferation and lead to increased neurogenesis [31]. However, there is no direct report showing the mechanism by which SFKs participate in NMDA receptors mediated neurogenesis after brain injury.

Advertisement

4. SFKs regulate cell cycle for mitogenic toxicity and cell proliferation after ICH

The cell cycle is an irreversible, ordered set of events that normally leads to cellular division [32-36]. The release of cells from a quiescent state (G0) results in their entry into the first gap phase (G1), during which the cells prepare for DNA replication in the synthetic phase (S). This is followed by the second gap phase (G2) and mitosis phase (M). After the cell has split into its two daughter cells, the new cells enter either G1 or G0. Mature neurons normally maintain themselves in G0 resting phase; however, a mature neuron that re-enters the cell cycle can neither advance to a new G0 quiescent state nor revert to its earlier G0 state. This presents a critical dilemma to the neuron from which death may be an unavoidable, but necessary, outcome for adult neurons attempting to complete the cell cycle [32,37]. Increasing evidence have revealed that aberrant cell cycle re-entry leads to neuronal death [8,32,37-64], and cell cycle inhibition via blocking SFKs can protect neurons from death post-ICH [8].

Apart from post-mitotic neurons, SFKs play critical roles in the process of cell cycle in dividable cells, by regulating mitogen-activated protein kinases (MAPKs) and cell cycle proteins such as cyclin-dependent kinases (Cdks) [65-69]. Although mitogenic signaling is necessary to initiate the cell cycle for normal cell division and proliferation, massive mitogenic signaling can also produce neurotoxicity and cell death [9,32,37,70]. Cell death and cell proliferation seem contradictory to each other, but these two seemingly different cellular processes share some common mitogenic molecules and signaling pathways (Fig. 1). In addition, many other molecules, including Ca2+, ROS, NO and MMPs can directly or indirectly activate or increase mitogenic signaling [54,71-77].

There are two stages (acute and recovery) after ICH (Fig. 1). Once ICH occurs, a large number of molecules (e.g., thrombin, glutamate, TNF-α, VEGF, etc.) are increased. This peaks within the first hour to a day in the acute stage after ICH, and then resolves gradually in the recovery stage after ICH. The over-activated SFKs/mitogenic signaling leads to neurons to enter the cell cycle and die, and damages astroctyes and BMVECs via MAPKs in acute stage after ICH. Within a day, however, the massive thrombin/SFK mitogenic signaling resolve, and the disease progresses to a recovery stage of ICH. The restored moderate SFK/mitogenic signaling leads to newborn BMVECs, astrocytes and other cells that mediate self-repair in the recovery stage after ICH.

As shown in Fig. 1, thrombin (a potent mitogen) triggers mitosis after ICH by modulating mitogenic intracellular molecules such as SFKs. SFKs participate in mitogenic signaling activation via regulating mitogen-activated protein kinases (MAPKs) and other molecules [64-69] that play critical roles not only in brain injuries during the acute stage in ICH, but in brain self-repair during the recovery stage in ICH. Acute inhibition of SFKs is beneficial, that attenuates hematoma, BBB breakdown, vasogenic edema, MAPK activation in the acute stage (0-24h) after ICH (Fig. 2, 3, 4, 5 & 6) [7-9,26,64,70]. In contrast, delayed and lasting inhibition of SFKs is detrimental, and prolongs BBB repair and brain edema resolution in the recovery stage (7-14 days) after ICH [9], presumably because SFKs mediate population of NPCs that exist in the “neurovascular niche”. that repair the damaged BBB [78]. Such NPCs could serve as a source of newborn cells (i.e., BMVECs, astrocytes and perhaps other cells) of the neurovascular unit that play a role in re-establishing the BBB via the mitogenic growth signaling pathways during recovery phase after ICH (Fig. 1) [79].

Figure 3.

Intracerebroventricular (i.c.v.) injection of thrombin (20 U/animal, i.c.v.) causes reductions in brain microvascular endothelial cell (BMVEC) immunoreactivity after 1 day, and subsequent BMVEC proliferation around the brain vessels in lacunosum moleculare layer (LMol) of the hippocampus after 7 days and 14 days in Sprague-Dawley rats. Panels A-C show rats following sham operations labeled for BrdU, bromodeoxyuridine (A), RECA-1, rat endothelial antigen-1 (B) and the overlay or Merged image (C). RECA-1+cells demonstrate the tube-shape of brain capillaries (arrows in panel C). Panels D to F show BrdU (D), RECA-1 (E) and the Merged image (F) at 1 day after thrombin injections. Compared with the sham group, RECA-1+cells tend to lose their tube-shape at 1 day following thrombin injections (arrows in panel F) and there were no BrdU+cells co-labeled with RECA-1 at one day (panel F). Panels G-I show the staining for BrdU (G), RECA-1 (H) and the Merged image (I) 1 day after thrombin plus PP2 injections. PP2 administration at day 0, immediately after thrombin injection, blocks the thrombin-induced loss of tube-shape of RECA-1+cells. Panels J-L show the staining of BrdU (J), RECA-1 (K) and the Merged image (L) 7 days after thrombin injection. Compared to 1 day, BrdU+cells are increased 7 days after thrombin injection. Some of these BrdU+cells are co-labeled with RECA-1 (arrows in panel L and M). A few brain capillaries regained their tube-shape, though not completely. Panel M shows a higher power image of Panel L (area within dashed lines). RECA-1 stained BMVEC are red. The BrdU+/RECA-1+double-labeled new born BMVEC nuclei are yellow. Panels N to P show the staining for BrdU (M), RECA-1 (N) and the Merged image (P) 14 days after thrombin injection. Compared to 7 days, BrdU+cells are decreased, but some BrdU+cells (N) remain co-labeled with RECA-1 (arrow in panel P), and more and more brain capillaries regained the tube-shape 14 days after the thrombin injection. Scale bars: A-P, 50 µm.

Figure 4.

Intracerebroventricular injection of thrombin (20 U/animal, i.c.v.) causes reductions in astrocyte glial fibrillary acidic protein (GFAP) immunoreactivity after 1 day, and subsequent astrocyte proliferation around the brain vessels in lacunosum moleculare layer (LMol) of the hippocampus after 7 days and 14 days in Sprague-Dawley rats. Panels A-C show rats with sham operation labeled for BrdU (A), GFAP (B), and Merged image (C). GFAP+cells envelop most all of the brain vessel (arrows in panel B). Panels D-F show BrdU (D), GFAP (E) and the Merged image (F) at 1 day after thrombin injection. Compared with the sham group, there is decreased GFAP immunoreactivity around brain vessels. There are a few BrdU+/GFAP-cells located close to the vessel (arrows in panel F). Panels G-I show the staining for BrdU (G), GFAP (H) and the Merged image (I) 1 day after thrombin plus PP2 injections. PP2 administration at day 0, immediately after thrombin injection, blocks the thrombin-induced reductions in GFAP immunoreactivity. Panels J-L show the staining for BrdU (J), GFAP (K) and the Merged image (L) 7 days after thrombin injection. Compared to 1 day, BrdU+cells are increased 7 days after thrombin injection (J, arrows). Many of these BrdU+cells are co-labeled with GFAP (arrows in panel L). Panel M shows a higher power image of Panel L (area within dashed lines). GFAP stained astrocytes are red. The BrdU+GFAP+double-labeled new born astrocytic nuclei are yellow (arrows, Panel M). Panels N-P show the staining for BrdU (N), GFAP (O) and the Merged image (P) 14 days after thrombin injection. Compared to 7 days, BrdU+cells are decreased 14 days after thrombin injection. Some BrdU+cells (N, arrow) remain co-labeled with GFAP (arrow in panel P) 14 days after the thrombin injection. Scale bars: A-P, 50 µm.

Figure 5.

Brain sodium fluorescein (NF, panel A) and Evans blue (EB, panel B) extravasation increased 1 day after thrombin (Throm) injections (20U, i.c.v.), and decreased at 7 and 14 days in Sprague-Dawley rats. The thrombin inhibitor hirudin (Hir, 20U) blocked thrombin-induced NF/EB extravasation at 1 day after co-injection into the cerebral ventricles. PP2 (src family kinase inhibitor) administered with thrombin (day 0) blocked the NF/EB extravasation at 1 day after thrombin injection, whereas delayed PP2 administration (days 2-6) postponed alleviation of NF/EB extravasation at 7 days post-thrombin injection. Each column and vertical bar represents the mean ± standard error of the mean. ** p<0.01 vs. Cont; #p<0.05, ##p<0.01 vs. Throm/1day, ‡‡ p<0.01 vs. Throm/7days (one-way ANOVA followed by Tukey’s post hoc test).

Figure 6.

Brain edema (water content) increased at 1 day after thrombin (Throm) injections (20U, i.c.v.), and decreased by 7 and 14 days in Sprague-Dawley rats. The thrombin inhibitor hirudin (Hir, 20U, i.c.v.) blocked elevation thrombin-induced brain water content at 1 day after co-injection into the cerebral ventricle. Administration of PP2 (src family kinase inhibitor) at day 0 blocked the increase in brain water content observed at 1 day after thrombin injection, whereas delayed PP2 administration (days 2-6) prevented the resolution of brain water content at 7 days post-thrombin injection. Each column and vertical bar represents the mean ± standard error of the mean. ** p<0.01 vs. Cont; #p<0.05, ##p<0.01 vs. Throm/1day, ‡ p<0.05 vs. Throm/7days (one-way ANOVA followed by Tukey’s post hoc test).

Advertisement

5. Future directions

Future studies need to address which specific SFK members found in brain (e.g., Src, Fyn, Lck and Yrk) that mediate ICH-induced cell death or birth. Since delayed and chronic inhibition of SFKs may impair neurogenesis and prolong BBB self-repair during recovery stage post-ICH, the acute and transient inhibition of SFKs should be pursued in treatment of ICH. The nanoparticle-based siRNA transfection system allows transient knockdown of target gene(s) and highly efficient delivery of siRNA in vivo with low cytotoxicity [80,81]. This could present a novel therapy for treating ICH patients as the nanoparticle-based siRNA approach provides heightened specificity for specific SFK gene(s) with less off target effects and this approach has been used in humans [82-85].

Advertisement

Acknowledgments

The authors acknowledge the support of AHA Beginning Grant-in-Aid 12BGIA12060381 (DZL) and NIH grant NS054652 (FRS). Figures 2, and 3-6 were published in Neurobiol Dis. 2008,30(2):201-11 and Ann Neurol. 2010,67(4):526-33, respectively.

References

  1. 1. Sudlow CL, Warlow CP (1997) Comparable studies of the incidence of stroke and its pathological types: results from an international collaboration. International Stroke Incidence Collaboration. Stroke 28: 491-499.
  2. 2. Flaherty ML, Haverbusch M, Sekar P, Kissela B, Kleindorfer D, et al. (2006) Long-term mortality after intracerebral hemorrhage. Neurology 66: 1182-1186.
  3. 3. Fogelholm R, Murros K, Rissanen A, Avikainen S (2005) Long term survival after primary intracerebral haemorrhage: a retrospective population based study. J Neurol Neurosurg Psychiatry 76: 1534-1538.
  4. 4. Gebel JM, Jr., Jauch EC, Brott TG, Khoury J, Sauerbeck L, et al. (2002) Natural history of perihematomal edema in patients with hyperacute spontaneous intracerebral hemorrhage. Stroke 33: 2631-2635.
  5. 5. Broderick J, Connolly S, Feldmann E, Hanley D, Kase C, et al. (2007) Guidelines for the management of spontaneous intracerebral hemorrhage in adults: 2007 update: a guideline from the American Heart Association/American Stroke Association Stroke Council, High Blood Pressure Research Council, and the Quality of Care and Outcomes in Research Interdisciplinary Working Group. Circulation 116: e391-413.
  6. 6. Ardizzone TD, Lu A, Wagner KR, Tang Y, Ran R, et al. (2004) Glutamate receptor blockade attenuates glucose hypermetabolism in perihematomal brain after experimental intracerebral hemorrhage in rat. Stroke 35: 2587-2591
  7. 7. Ardizzone TD, Zhan X, Ander BP, Sharp FR (2007) SRC kinase inhibition improves acute outcomes after experimental intracerebral hemorrhage. Stroke 38: 1621-1625.
  8. 8. Liu DZ, Cheng XY, Ander BP, Xu H, Davis RR, et al. (2008) Src kinase inhibition decreases thrombin-induced injury and cell cycle re-entry in striatal neurons. Neurobiol Dis 30: 201-211.
  9. 9. Liu DZ, Ander BP, Xu H, Shen Y, Kaur P, et al. (2010) Blood-brain barrier breakdown and repair by Src after thrombin-induced injury. Ann Neurol 67: 526-533.
  10. 10. Oda H, Kumar S, Howley PM (1999) Regulation of the Src family tyrosine kinase Blk through E6AP-mediated ubiquitination. Proc Natl Acad Sci U S A 96: 9557-9562.
  11. 11. Biscardi JS, Ishizawar RC, Silva CM, Parsons SJ (2000) Tyrosine kinase signalling in breast cancer: epidermal growth factor receptor and c-Src interactions in breast cancer. Breast Cancer Res 2: 203-210.
  12. 12. Boggon TJ, Eck MJ (2004) Structure and regulation of Src family kinases. Oncogene 23: 7918-7927.
  13. 13. Parsons SJ, Parsons JT (2004) Src family kinases, key regulators of signal transduction. Oncogene 23: 7906-7909.
  14. 14. Morse WR, Whitesides JG, 3rd, LaMantia AS, Maness PF (1998) p59fyn and pp60c-src modulate axonal guidance in the developing mouse olfactory pathway. J Neurobiol 36: 53-63.
  15. 15. Encinas M, Tansey MG, Tsui-Pierchala BA, Comella JX, Milbrandt J, et al. (2001) c-Src is required for glial cell line-derived neurotrophic factor (GDNF) family ligand-mediated neuronal survival via a phosphatidylinositol-3 kinase (PI-3K)-dependent pathway. J Neurosci 21: 1464-1472.
  16. 16. Sperber BR, Boyle-Walsh EA, Engleka MJ, Gadue P, Peterson AC, et al. (2001) A unique role for Fyn in CNS myelination. J Neurosci 21: 2039-2047.
  17. 17. Heidinger V, Manzerra P, Wang XQ, Strasser U, Yu SP, et al. (2002) Metabotropic glutamate receptor 1-induced upregulation of NMDA receptor current: mediation through the Pyk2/Src-family kinase pathway in cortical neurons. J Neurosci 22: 5452-5461.
  18. 18. Rouer E (2010) [Neuronal isoforms of Src, Fyn and Lck tyrosine kinases: A specific role for p56lckN in neuron protection]. C R Biol 333: 1-10.
  19. 19. Salter MW, Kalia LV (2004) Src kinases: a hub for NMDA receptor regulation. Nat Rev Neurosci 5: 317-328.
  20. 20. Stein PL, Vogel H, Soriano P (1994) Combined deficiencies of Src, Fyn, and Yes tyrosine kinases in mutant mice. Genes Dev 8: 1999-2007.
  21. 21. Hunter T (1987) A tail of two src's: mutatis mutandis. Cell 49: 1-4.
  22. 22. McBain CJ, Mayer ML (1994) N-methyl-D-aspartic acid receptor structure and function. Physiol Rev 74: 723-760.
  23. 23. Frank RA (2011) Endogenous ion channel complexes: the NMDA receptor. Biochem Soc Trans 39: 707-718.
  24. 24. Ali DW, Salter MW (2001) NMDA receptor regulation by Src kinase signalling in excitatory synaptic transmission and plasticity. Curr Opin Neurobiol 11: 336-342.
  25. 25. Groveman BR, Feng S, Fang XQ, Pflueger M, Lin SX, et al. (2012) The regulation of N-methyl-D-aspartate receptors by Src kinase. FEBS J 279: 20-28.
  26. 26. Sharp F, Liu DZ, Zhan X, Ander BP (2008) Intracerebral hemorrhage injury mechanisms: glutamate neurotoxicity, thrombin, and Src. Acta Neurochir Suppl 105: 43-46.
  27. 27. Hossain MI, Kamaruddin MA, Cheng HC (2012) Aberrant regulation and function of Src family tyrosine kinases: their potential contributions to glutamate-induced neurotoxicity. Clin Exp Pharmacol Physiol 39: 684-691.
  28. 28. Yang M, Leonard JP (2001) Identification of mouse NMDA receptor subunit NR2A C-terminal tyrosine sites phosphorylated by coexpression with v-Src. J Neurochem 77: 580-588.
  29. 29. Zheng F, Gingrich MB, Traynelis SF, Conn PJ (1998) Tyrosine kinase potentiates NMDA receptor currents by reducing tonic zinc inhibition. Nat Neurosci 1: 185-191.
  30. 30. Kohr G, Seeburg PH (1996) Subtype-specific regulation of recombinant NMDA receptor-channels by protein tyrosine kinases of the src family. J Physiol 492 (Pt 2): 445-452.
  31. 31. Suzuki M, Nelson AD, Eickstaedt JB, Wallace K, Wright LS, et al. (2006) Glutamate enhances proliferation and neurogenesis in human neural progenitor cell cultures derived from the fetal cortex. Eur J Neurosci 24: 645-653.
  32. 32. Liu DZ, Ander BP, Sharp FR (2010) Cell cycle inhibition without disruption of neurogenesis is a strategy for treatment of central nervous system diseases. Neurobiol Dis 37: 549-557.
  33. 33. Novak B, Tyson JJ, Gyorffy B, Csikasz-Nagy A (2007) Irreversible cell-cycle transitions are due to systems-level feedback. Nat Cell Biol 9: 724-728.
  34. 34. Norbury C, Nurse P (1992) Animal cell cycles and their control. Annu Rev Biochem 61: 441-470.
  35. 35. Malumbres M, Barbacid M (2001) To cycle or not to cycle: a critical decision in cancer. Nat Rev Cancer 1: 222-231.
  36. 36. Schwartz GK, Shah MA (2005) Targeting the cell cycle: a new approach to cancer therapy. J Clin Oncol 23: 9408-9421.
  37. 37. Liu DZ, Ander BP (2012) Cell cycle inhibition without disruption of neurogenesis is a strategy for treatment of aberrant cell cycle diseases: an update. ScientificWorldJournal 2012: 491737.
  38. 38. Nagy Z, Esiri MM, Cato AM, Smith AD (1997) Cell cycle markers in the hippocampus in Alzheimer's disease. Acta Neuropathol 94: 6-15.
  39. 39. McShea A, Harris PL, Webster KR, Wahl AF, Smith MA (1997) Abnormal expression of the cell cycle regulators P16 and CDK4 in Alzheimer's disease. Am J Pathol 150: 1933-1939.
  40. 40. Nagy Z, Esiri MM (1998) Neuronal cyclin expression in the hippocampus in temporal lobe epilepsy. Exp Neurol 150: 240-247.
  41. 41. Busser J, Geldmacher DS, Herrup K (1998) Ectopic cell cycle proteins predict the sites of neuronal cell death in Alzheimer's disease brain. J Neurosci 18: 2801-2807.
  42. 42. Copani A, Condorelli F, Caruso A, Vancheri C, Sala A, et al. (1999) Mitotic signaling by beta-amyloid causes neuronal death. Faseb J 13: 2225-2234.
  43. 43. Yang Y, Geldmacher DS, Herrup K (2001) DNA replication precedes neuronal cell death in Alzheimer's disease. J Neurosci 21: 2661-2668.
  44. 44. Copani A, Uberti D, Sortino MA, Bruno V, Nicoletti F, et al. (2001) Activation of cell-cycle-associated proteins in neuronal death: a mandatory or dispensable path? Trends Neurosci 24: 25-31.
  45. 45. O'Hare M, Wang F, Park DS (2002) Cyclin-dependent kinases as potential targets to improve stroke outcome. Pharmacol Ther 93: 135-143.
  46. 46. Lefevre K, Clarke PG, Danthe EE, Castagne V (2002) Involvement of cyclin-dependent kinases in axotomy-induced retinal ganglion cell death. J Comp Neurol 447: 72-81.
  47. 47. Imai H, Harland J, McCulloch J, Graham DI, Brown SM, et al. (2002) Specific expression of the cell cycle regulation proteins, GADD34 and PCNA, in the peri-infarct zone after focal cerebral ischaemia in the rat. Eur J Neurosci 15: 1929-1936.
  48. 48. Jordan-Sciutto KL, Dorsey R, Chalovich EM, Hammond RR, Achim CL (2003) Expression patterns of retinoblastoma protein in Parkinson disease. J Neuropathol Exp Neurol 62: 68-74.
  49. 49. Yang Y, Mufson EJ, Herrup K (2003) Neuronal cell death is preceded by cell cycle events at all stages of Alzheimer's disease. J Neurosci 23: 2557-2563.
  50. 50. Ranganathan S, Bowser R (2003) Alterations in G(1) to S phase cell-cycle regulators during amyotrophic lateral sclerosis. Am J Pathol 162: 823-835.
  51. 51. Nguyen MD, Boudreau M, Kriz J, Couillard-Despres S, Kaplan DR, et al. (2003) Cell cycle regulators in the neuronal death pathway of amyotrophic lateral sclerosis caused by mutant superoxide dismutase 1. J Neurosci 23: 2131-2140.
  52. 52. Herrup K, Neve R, Ackerman SL, Copani A (2004) Divide and die: cell cycle events as triggers of nerve cell death. J Neurosci 24: 9232-9239.
  53. 53. Kuan CY, Schloemer AJ, Lu A, Burns KA, Weng WL, et al. (2004) Hypoxia-ischemia induces DNA synthesis without cell proliferation in dying neurons in adult rodent brain. J Neurosci 24: 10763-10772.
  54. 54. Copani A, Nicoletti F (2005) Cell-cycle mechanisms and neuronal cell death. New York: Kluwer Academic/Plenum.
  55. 55. Di Giovanni S, Movsesyan V, Ahmed F, Cernak I, Schinelli S, et al. (2005) Cell cycle inhibition provides neuroprotection and reduces glial proliferation and scar formation after traumatic brain injury. Proc Natl Acad Sci U S A 102: 8333-8338.
  56. 56. Andorfer C, Acker CM, Kress Y, Hof PR, Duff K, et al. (2005) Cell-cycle reentry and cell death in transgenic mice expressing nonmutant human tau isoforms. J Neurosci 25: 5446-5454.
  57. 57. Copani A, Hoozemans JJ, Caraci F, Calafiore M, Van Haastert ES, et al. (2006) DNA polymerase-beta is expressed early in neurons of Alzheimer's disease brain and is loaded into DNA replication forks in neurons challenged with beta-amyloid. J Neurosci 26: 10949-10957.
  58. 58. Zhang Y, Qu D, Morris EJ, O'Hare MJ, Callaghan SM, et al. (2006) The Chk1/Cdc25A pathway as activators of the cell cycle in neuronal death induced by camptothecin. J Neurosci 26: 8819-8828.
  59. 59. Yang Y, Varvel NH, Lamb BT, Herrup K (2006) Ectopic cell cycle events link human Alzheimer's disease and amyloid precursor protein transgenic mouse models. J Neurosci 26: 775-784.
  60. 60. Khurana V, Lu Y, Steinhilb ML, Oldham S, Shulman JM, et al. (2006) TOR-mediated cell-cycle activation causes neurodegeneration in a Drosophila tauopathy model. Curr Biol 16: 230-241.
  61. 61. Rao HV, Thirumangalakudi L, Desmond P, Grammas P (2007) Cyclin D1, cdk4, and Bim are involved in thrombin-induced apoptosis in cultured cortical neurons. J Neurochem 101: 498-505.
  62. 62. Zhang J, Herrup K (2011) Nucleocytoplasmic Cdk5 is involved in neuronal cell cycle and death in post-mitotic neurons. Cell Cycle 10: 1208-1214.
  63. 63. Liu DZ, Ander BP (2011) Cell cycle phase transitions: signposts for aberrant cell cycle reentry in dying mature neurons. Journal of Cytology & Histology 2: 5.
  64. 64. (2004) Finishing the euchromatic sequence of the human genome. Nature 431: 931-945.
  65. 65. Grimmler M, Wang Y, Mund T, Cilensek Z, Keidel EM, et al. (2007) Cdk-inhibitory activity and stability of p27Kip1 are directly regulated by oncogenic tyrosine kinases. Cell 128: 269-280.
  66. 66. Kasahara K, Nakayama Y, Nakazato Y, Ikeda K, Kuga T, et al. (2007) Src signaling regulates completion of abscission in cytokinesis through ERK/MAPK activation at the midbody. J Biol Chem 282: 5327-5339.
  67. 67. Liu Z, Falola J, Zhu X, Gu Y, Kim LT, et al. (2004) Antiproliferative effects of Src inhibition on medullary thyroid cancer. J Clin Endocrinol Metab 89: 3503-3509.
  68. 68. Mishra R, Wang Y, Simonson MS (2005) Cell cycle signaling by endothelin-1 requires Src nonreceptor protein tyrosine kinase. Mol Pharmacol 67: 2049-2056.
  69. 69. Taylor SJ, Shalloway D (1993) The cell cycle and c-Src. Curr Opin Genet Dev 3: 26-34.
  70. 70. Liu DZ, Sharp FR (2011) The dual role of SRC kinases in intracerebral hemorrhage. Acta Neurochir Suppl 111: 77-81.
  71. 71. Li N, Wang C, Wu Y, Liu X, Cao X (2009) Ca2+/Calmodulin-dependent Protein Kinase II Promotes Cell Cycle Progression by Directly Activating MEK1 and Subsequently Modulating p27 Phosphorylation. J Biol Chem 284: 3021-3027.
  72. 72. Mao LM, Tang QS, Wang JQ (2009) Regulation of extracellular signal-regulated kinase phosphorylation in cultured rat striatal neurons. Brain Res Bull 78: 328-334.
  73. 73. de Bernardo S, Canals S, Casarejos MJ, Solano RM, Menendez J, et al. (2004) Role of extracellular signal-regulated protein kinase in neuronal cell death induced by glutathione depletion in neuron/glia mesencephalic cultures. J Neurochem 91: 667-682.
  74. 74. Qian JY, Leung A, Harding P, LaPointe MC (2006) PGE2 stimulates human brain natriuretic peptide expression via EP4 and p42/44 MAPK. Am J Physiol Heart Circ Physiol 290: H1740-1746.
  75. 75. Fiebich BL, Schleicher S, Spleiss O, Czygan M, Hull M (2001) Mechanisms of prostaglandin E2-induced interleukin-6 release in astrocytes: possible involvement of EP4-like receptors, p38 mitogen-activated protein kinase and protein kinase C. J Neurochem 79: 950-958.
  76. 76. Meissirel C, Ruiz de Almodovar C, Knevels E, Coulon C, Chounlamountri N, et al. (2011) VEGF modulates NMDA receptors activity in cerebellar granule cells through Src-family kinases before synapse formation. Proc Natl Acad Sci U S A 108: 13782-13787.
  77. 77. Rosenberg GA (2009) Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol 8: 205-216.
  78. 78. Ohab JJ, Fleming S, Blesch A, Carmichael ST (2006) A neurovascular niche for neurogenesis after stroke. J Neurosci 26: 13007-13016.
  79. 79. Palmer TD, Willhoite AR, Gage FH (2000) Vascular niche for adult hippocampal neurogenesis. J Comp Neurol 425: 479-494.
  80. 80. Hannon GJ (2002) RNA interference. Nature 418: 244-251.
  81. 81. Reynolds A, Leake D, Boese Q, Scaringe S, Marshall WS, et al. (2004) Rational siRNA design for RNA interference. Nat Biotechnol 22: 326-330.
  82. 82. de Fougerolles A, Vornlocher HP, Maraganore J, Lieberman J (2007) Interfering with disease: a progress report on siRNA-based therapeutics. Nat Rev Drug Discov 6: 443-453.
  83. 83. Eifler AC, Thaxton CS (2011) Nanoparticle therapeutics: FDA approval, clinical trials, regulatory pathways, and case study. Methods Mol Biol 726: 325-338.
  84. 84. Davis ME (2009) The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol Pharm 6: 659-668.
  85. 85. Davis ME, Zuckerman JE, Choi CH, Seligson D, Tolcher A, et al. (2010) Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 464: 1067-1070.

Written By

DaZhi Liu

Submitted: 11 September 2013 Published: 10 September 2014