Neuroprotective pentapeptide CN-105 is associated with reduced sterile inflammation and improved functional outcomes in a traumatic brain injury murine model

Daniel T Laskowitz, Haichen Wang, Tony Chen, David T Lubkin, Viviana Cantillana, Tian Ming Tu, Dawn Kernagis, Guanen Zhou, Gary Macy, Bradley J Kolls, Hana N Dawson, Daniel T Laskowitz, Haichen Wang, Tony Chen, David T Lubkin, Viviana Cantillana, Tian Ming Tu, Dawn Kernagis, Guanen Zhou, Gary Macy, Bradley J Kolls, Hana N Dawson

Abstract

At present, there are no proven pharmacological treatments demonstrated to improve long term functional outcomes following traumatic brain injury(TBI). In the setting of non-penetrating TBI, sterile brain inflammatory responses are associated with the development of cerebral edema, intracranial hypertension, and secondary neuronal injury. There is increasing evidence that endogenous apolipoprotein E(apoE) modifies the neuroinflammatory response through its role in downregulating glial activation, however, the intact apoE holoprotein does not cross the blood-brain barrier due to its size. To address this limitation, we developed a small 5 amino acid apoE mimetic peptide(CN-105) that mimics the polar face of the apoE helical domain involved in receptor interactions. The goal of this study was to investigate the therapeutic potential of CN-105 in a murine model of closed head injury. Treatment with CN-105 was associated with a durable improvement in functional outcomes as assessed by Rotarod and Morris Water Maze and a reduction in positive Fluoro-Jade B stained injured neurons and microglial activation. Administration of CN-105 was also associated with reduction in mRNA expression of a subset of inflammatory and immune-related genes.

Conflict of interest statement

The patent for CN-105 is held by Duke University, and D.T. La, B.J.K., and H.N.D. are co-inventors. D.T. La is an officer of Aegis-CN, which supplied drug for these studies, but had no role in experimental design or writing of this manuscript.

Figures

Figure 1. CN-105 improves vestibulomor and cognitive…
Figure 1. CN-105 improves vestibulomor and cognitive function.
Following TBI, intravenous administration of 0.05 mg/kg of CN-105 was the lowest effective dose resulting in improved vestibulomotor functional performance, as assessed by Rotarod(A) and cognitive function as assessed by the Morris Water Maze(B) and the MWM probe trial(C). There was no difference in swim speed on the MWM(D). CN-105(0.05 mg/kg) administered 4 hours post-injury was also associated with a significant and durable improvement in Rotarod latency(E) and by reduced deficit on MWM(F) and the MWM probe trial(G) No significant difference in speed was detected between the two treatment groups(H). For comparison purposes results from sham mice are shown(dotted lines) in panels A and B but are not included in statistical calculations. Asterisks denote significant differences in performance as measured by ANOVA; *p < 0.05 and ** p < 0.01.
Figure 2. CN-105 reduces microgliosis and neuronal…
Figure 2. CN-105 reduces microgliosis and neuronal injury.
Comparison images of activated F4/80 immunostained microglial in the hippocampus 10 days post-TBI, brain slices from vehicle(A,C,E,G) and CN-105(B,D,F,H) treated mice, unbiased stereology confirms that treatment with CN-105 is associated with a reduction in microgliosis(p = 0.0002, ***)(I). Higher powered images of microglia in the CA3 region(C,D) in the polymorphic region(E,F) and in corpus callosum and fimbria of the periventricular region(G,H). Microglia are indicated in with arrows and phagocytic microglia with arrowheads in E and F. Images C-H are at 20x magnification and the black bar in G represents 40 μm. Images A and B are at 4x magnification and the white bar in A represents 250 μm. Comparison images of FJB-stained brain slices 24 hours post-TBI show degenerating neurons in the hilus of the dorsal dentate gyrus of TBI vehicle treated mice(J) which are significantly reduced by treatment with CN-105(K), quantification p < 0.0001(L).
Figure 3. CN-105 ameliorates changes in inflammatory…
Figure 3. CN-105 ameliorates changes in inflammatory gene expression.
One day post-injury, differential inflammatory gene expression analysis demonstrated changes in gene expression relative to sham vehicle treated controls for TBI CN-105 treated and untreated mice. Results are organized by upregulated(A) and downregulated(B) expression of genes from vehicle treated TBI mice relative to vehicle treated sham mice. Results from CN-105 treated TBI and sham mice relative to vehicle treated sham mice are also included. Only genes from TBI CN-105 treated mice that showed more than a 3 fold change when compared to TBI vehicle treated mice are presented.
Figure 4. Pharmacokinetic studies of CN-105 demonstrate…
Figure 4. Pharmacokinetic studies of CN-105 demonstrate CNS bioavailability.
Concentration of [14C]- radioactivity in the plasma and central nervous system of male CD-1 mice following an intravenous dose of [14C]-radiolabeled CN-105 peptide(A). For clarity, the concentration of [14C]- radioactivity during the first 6 hours only is depicted in(B).
Figure 5. TLR signaling is downregulated by…
Figure 5. TLR signaling is downregulated by CN-105.
(A) A schematic of the signaling pathway proposed for sterile inflammation post injury. Green arrows indicate upregulation and red arrows downregulation of genes 24 hours after TBI.(B) Graphic representation of genes 24 hours post TBI demonstrating fold change compared to sham control.(C) CN-105 modifies the post-injury inflammatory response. Arrows indicate reduced upregulation(green) and reduced downregulation(red) of gene expression when TBI injured mice are treated with CN-105 as compared to vehicle.(D) Graphic representation of differential gene expression 24 hours post TBI demonstrating fold change in gene expression of CN-105 treated mice normalized to vehicle treated TBI mice.

References

    1. Faul M., Xu L., Wald M. M., Coronado V. & Dellinger A. M. Traumatic Brain Injury in the United States: National Estimates of Prevalence and Incidence, 2002–2006. Injury Prev 16, A268–A268, doi: 10.1136/ip.2010.029951(2010).
    1. Flanagan S. R. Invited Commentary on “Centers for Disease Control and Prevention Report to Congress: Traumatic Brain Injury in the United States: Epidemiology and Rehabilitation”. Arch Phys Med Rehab 96, 1753–1755, doi: 10.1016/j.apmr.2015.07.001(2015).
    1. Selassie A. W. et al.. Incidence of long-term disability following traumatic brain injury hospitalization, United States, 2003. J Head Trauma Rehab 23, 123–131, doi: 10.1097/01.Htr.0000314531.30401.39(2008).
    1. Zaloshnja E., Miller T., Langlois J. A. & Selassie A. W. Prevalence of Long-Term Disability From Traumatic Brain Injury in the Civilian Population of the United States, 2005. J Head Trauma Rehab 23, 394–400, doi: 10.1097/(2008).
    1. Corps K. N., Roth T. L. & McGavern D. B. Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol 72, 355–362, doi: 10.1001/jamaneurol.2014.3558(2015).
    1. Roberts I. et al.. Effect of intravenous corticosteroids on death within 14 days in 10008 adults with clinically significant head injury(MRC CRASH trial): randomised placebo-controlled trial. Lancet 364, 1321–1328, doi: 10.1016/S0140-6736(04)17188-2(2004).
    1. McConeghy K. W., Hatton J., Hughes L. & Cook A. M. A review of neuroprotection pharmacology and therapies in patients with acute traumatic brain injury. CNS drugs 26, 613–636, doi: 10.2165/11634020-000000000-00000(2012).
    1. Lanterna L. A. et al.. Meta-analysis of APOE genotype and subarachnoid hemorrhage: clinical outcome and delayed ischemia. Neurology 69, 766–775, doi: 10.1212/01.wnl.0000267640.03300.6b(2007).
    1. Morris P. G., Wilson J. T., Dunn L. T. & Nicoll J. A. Apolipoprotein E polymorphism and neuropsychological outcome following subarachnoid haemorrhage. Acta Neurol Scand 109, 205–209(2004).
    1. Alberts M. J. et al.. ApoE genotype and survival from intracerebral haemorrhage. Lancet 346, 575(1995).
    1. Biffi A. et al.. APOE genotype and extent of bleeding and outcome in lobar intracerebral haemorrhage: a genetic association study. Lancet Neurol 10, 702–709, doi: 10.1016/S1474-4422(11)70148-X(2011).
    1. Alexander S. et al.. Apolipoprotein E4 allele presence and functional outcome after severe traumatic brain injury. Journal of neurotrauma 24, 790–797, doi: 10.1089/neu.2006.0133(2007).
    1. Kassam I., Gagnon F. & Cusimano M. D. Association of the APOE-epsilon4 allele with outcome of traumatic brain injury in children and youth: a meta-analysis and meta-regression. J Neurol Neurosurg Psychiatry 87, 433–440, doi: 10.1136/jnnp-2015-310500(2016).
    1. Lawrence D. W., Comper P., Hutchison M. G. & Sharma B. The role of apolipoprotein E episilon(epsilon)-4 allele on outcome following traumatic brain injury: A systematic review. Brain Inj 29, 1018–1031, doi: 10.3109/02699052.2015.1005131(2015).
    1. Li L. et al.. The Association Between Apolipoprotein E and Functional Outcome After Traumatic Brain Injury: A Meta-Analysis. Medicine(Baltimore) 94, e2028, doi: 10.1097/MD.0000000000002028(2015).
    1. Willemse-van Son A. H., Ribbers G. M., Hop W. C., van Duijn C. M. & Stam H. J. Association between apolipoprotein-epsilon4 and long-term outcome after traumatic brain injury. J Neurol Neurosurg Psychiatry 79, 426–430, doi: 10.1136/jnnp.2007.129460(2008).
    1. Zhou W. et al.. Meta-analysis of APOE4 allele and outcome after traumatic brain injury. Journal of neurotrauma 25, 279–290, doi: 10.1089/neu.2007.0489(2008).
    1. Laskowitz D. T., Goel S., Bennett E. R. & Matthew W. D. Apolipoprotein E suppresses glial cell secretion of TNF alpha. Journal of neuroimmunology 76, 70–74(1997).
    1. Lynch J. R. et al.. Apolipoprotein E affects the central nervous system response to injury and the development of cerebral edema. Annals of neurology 51, 113–117(2002).
    1. Linton M. F. et al.. Phenotypes of apolipoprotein B and apolipoprotein E after liver transplantation. The Journal of clinical investigation 88, 270–281, doi: 10.1172/JCI115288(1991).
    1. Laskowitz D. T., Fillit H., Yeung N., Toku K. & Vitek M. P. Apolipoprotein E-derived peptides reduce CNS inflammation: implications for therapy of neurological disease. Acta neurologica Scandinavica. Supplementum 185, 15–20, doi: 10.1111/j.1600-0404.2006.00680.x(2006).
    1. Laskowitz D. T. et al.. Downregulation of microglial activation by apolipoprotein E and apoE-mimetic peptides. Experimental neurology 167, 74–85, doi: 10.1006/exnr.2001.7541(2001).
    1. Lynch J. R. et al.. APOE genotype and an ApoE-mimetic peptide modify the systemic and central nervous system inflammatory response. The Journal of biological chemistry 278, 48529–48533, doi: 10.1074/jbc.M306923200(2003).
    1. James M. L., Sullivan P. M., Lascola C. D., Vitek M. P. & Laskowitz D. T. Pharmacogenomic effects of apolipoprotein e on intracerebral hemorrhage. Stroke; a journal of cerebral circulation 40, 632–639, doi: 10.1161/STROKEAHA.108.530402(2009).
    1. Gao J. et al.. A novel apoE-derived therapeutic reduces vasospasm and improves outcome in a murine model of subarachnoid hemorrhage. Neurocritical care 4, 25–31, doi: 10.1385/NCC:4:1:025(2006).
    1. Mesis R. G. et al.. Dissociation between vasospasm and functional improvement in a murine model of subarachnoid hemorrhage. Neurosurgical focus 21, E4(2006).
    1. Hoane M. R. et al.. The novel apolipoprotein E-based peptide COG1410 improves sensorimotor performance and reduces injury magnitude following cortical contusion injury. Journal of neurotrauma 24, 1108–1118, doi: 10.1089/neu.2006.0254(2007).
    1. Hoane M. R., Kaufman N., Vitek M. P. & McKenna S. E. COG1410 improves cognitive performance and reduces cortical neuronal loss in the traumatically injured brain. Journal of neurotrauma 26, 121–129, doi: 10.1089/neu.2008.0565(2009).
    1. Laskowitz D. T. et al.. Traumatic brain injury exacerbates neurodegenerative pathology: improvement with an apolipoprotein E-based therapeutic. Journal of neurotrauma 27, 1983–1995, doi: 10.1089/neu.2010.1396(2010).
    1. Lynch J. R. et al.. A novel therapeutic derived from apolipoprotein E reduces brain inflammation and improves outcome after closed head injury. Experimental neurology 192, 109–116, doi: 10.1016/j.expneurol.2004.11.014(2005).
    1. Hoe H. S., Harris D. C. & Rebeck G. W. Multiple pathways of apolipoprotein E signaling in primary neurons. Journal of neurochemistry 93, 145–155, doi: 10.1111/j.1471-4159.2004.03007.x(2005).
    1. Misra U. K. et al.. Apolipoprotein E and mimetic peptide initiate a calcium-dependent signaling response in macrophages. Journal of leukocyte biology 70, 677–683(2001).
    1. Pocivavsek A., Burns M. P. & Rebeck G. W. Low-density lipoprotein receptors regulate microglial inflammation through c-Jun N-terminal kinase. Glia 57, 444–453, doi: 10.1002/glia.20772(2009).
    1. Pocivavsek A., Mikhailenko I., Strickland D. K. & Rebeck G. W. Microglial low-density lipoprotein receptor-related protein 1 modulates c-Jun N-terminal kinase activation. Journal of neuroimmunology 214, 25–32, doi: 10.1016/j.jneuroim.2009.06.010(2009).
    1. Cernak I. et al.. The pathobiology of moderate diffuse traumatic brain injury as identified using a new experimental model of injury in rats. Neurobiology of disease 17, 29–43, doi: 10.1016/j.nbd.2004.05.011(2004).
    1. Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. Journal of neuroscience methods 11, 47–60(1984).
    1. Yu T. S., Zhang G., Liebl D. J. & Kernie S. G. Traumatic brain injury-induced hippocampal neurogenesis requires activation of early nestin-expressing progenitors. The Journal of neuroscience: the official journal of the Society for Neuroscience 28, 12901–12912, doi: 10.1523/JNEUROSCI.4629-08.2008(2008).
    1. Iyer S. S. & Cheng G. Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit Rev Immunol 32, 23–63(2012).
    1. Yuan X., Peng X., Li Y. & Li M. Role of IL-38 and its related cytokines in inflammation. Mediators Inflamm 2015, 807976, doi: 10.1155/2015/807976(2015).
    1. van de Veerdonk F. L. et al.. IL-38 binds to the IL-36 receptor and has biological effects on immune cells similar to IL-36 receptor antagonist. Proceedings of the National Academy of Sciences of the United States of America 109, 3001–3005, doi: 10.1073/pnas.1121534109(2012).
    1. Mora J. et al.. Interleukin-38 is released from apoptotic cells to limit inflammatory macrophage responses. J Mol Cell Biol, doi: 10.1093/jmcb/mjw006(2016).
    1. Can A. et al.. Antidepressant-like responses to lithium in genetically diverse mouse strains. Genes Brain Behav 10, 434–443, doi: 10.1111/j.1601-183X.2011.00682.x(2011).
    1. Macleod J. N., Sorensen M. P. & Shapiro B. H. Strain independent elevation of hepatic mono-oxygenase enzymes in female mice. Xenobiotica 17, 1095–1102(1987).
    1. Han S. H. & Chung S. Y. Marked hippocampal neuronal damage without motor deficits after mild concussive-like brain injury in apolipoprotein E-deficient mice. Annals of the New York Academy of Sciences 903, 357–365(2000).
    1. Aono M. et al.. Protective effect of apolipoprotein E-mimetic peptides on N-methyl-D-aspartate excitotoxicity in primary rat neuronal-glial cell cultures. Neuroscience 116, 437–445(2003).
    1. Laskowitz D. T. et al.. COG1410, a novel apolipoprotein E-based peptide, improves functional recovery in a murine model of traumatic brain injury. Journal of neurotrauma 24, 1093–1107, doi: 10.1089/neu.2006.0192(2007).
    1. Qiu Z., Crutcher K. A., Hyman B. T. & Rebeck G. W. ApoE isoforms affect neuronal N-methyl-D-aspartate calcium responses and toxicity via receptor-mediated processes. Neuroscience 122, 291–303(2003).
    1. Sheng Z., Prorok M., Brown B. E. & Castellino F. J. N-methyl-D-aspartate receptor inhibition by an apolipoprotein E-derived peptide relies on low-density lipoprotein receptor-associated protein. Neuropharmacology 55, 204–214, doi: 10.1016/j.neuropharm.2008.05.016(2008).
    1. Fan L. et al.. Experimental brain injury induces differential expression of tumor necrosis factor-alpha mRNA in the CNS. Brain Res Mol Brain Res 36, 287–291(1996).
    1. Kinoshita K. et al.. Interleukin-1beta messenger ribonucleic acid and protein levels after fluid-percussion brain injury in rats: importance of injury severity and brain temperature. Neurosurgery 51, 195–203, discussion 203(2002).
    1. Szmydynger-Chodobska J., Strazielle N., Zink B. J., Ghersi-Egea J. F. & Chodobski A. The role of the choroid plexus in neutrophil invasion after traumatic brain injury. J Cereb Blood Flow Metab 29, 1503–1516, doi: 10.1038/jcbfm.2009.71(2009).
    1. Bergold P. J. Treatment of traumatic brain injury with anti-inflammatory drugs. Experimental neurology 275 Pt 3, 367–380, doi: 10.1016/j.expneurol.2015.05.024(2016).
    1. Woodcock T. & Morganti-Kossmann M. C. The role of markers of inflammation in traumatic brain injury. Front Neurol 4, 18, doi: 10.3389/fneur.2013.00018(2013).
    1. Longhi L. et al.. Effect of traumatic brain injury on cognitive function in mice lacking p55 and p75 tumor necrosis factor receptors. Acta Neurochir Suppl 102, 409–413(2008).
    1. Scherbel U. et al.. Differential acute and chronic responses of tumor necrosis factor-deficient mice to experimental brain injury. Proceedings of the National Academy of Sciences of the United States of America 96, 8721–8726(1999).
    1. Stahel P. F. et al.. Experimental closed head injury: analysis of neurological outcome, blood-brain barrier dysfunction, intracranial neutrophil infiltration, and neuronal cell death in mice deficient in genes for pro-inflammatory cytokines. J Cereb Blood Flow Metab 20, 369–380, doi: 10.1097/00004647-200002000-00019(2000).
    1. Shohami E., Ginis I. & Hallenbeck J. M. Dual role of tumor necrosis factor alpha in brain injury. Cytokine & growth factor reviews 10, 119–130(1999).
    1. Ziebell J. M. & Morganti-Kossmann M. C. Involvement of pro- and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury. Neurotherapeutics 7, 22–30, doi: 10.1016/j.nurt.2009.10.016(2010).
    1. Li G. Z. et al.. Expression of myeloid differentiation primary response protein 88(Myd88) in the cerebral cortex after experimental traumatic brain injury in rats. Brain Res 1396, 96–104, doi: 10.1016/j.brainres.2011.04.014(2011).
    1. Li W. et al.. Enhanced cortical expression of myeloid differentiation primary response protein 88(Myd88) in patients with traumatic brain injury. J Surg Res 180, 133–139, doi: 10.1016/j.jss.2012.10.928(2013).
    1. Koedel U. et al.. Acute brain injury triggers MyD88-dependent, TLR2/4-independent inflammatory responses. The American journal of pathology 171, 200–213, doi: 10.2353/ajpath.2007.060821(2007).
    1. Gonias S. L. & Campana W. M. LDL receptor-related protein-1: a regulator of inflammation in atherosclerosis, cancer, and injury to the nervous system. The American journal of pathology 184, 18–27, doi: 10.1016/j.ajpath.2013.08.029(2014).
    1. Bell R. D. et al.. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 485, 512–516, doi: 10.1038/nature11087(2012).
    1. Croy J. E., Brandon T. & Komives E. A. Two apolipoprotein E mimetic peptides, ApoE(130-149) and ApoE(141-155)2, bind to LRP1. Biochemistry 43, 7328–7335, doi: 10.1021/bi036208p(2004).
    1. Ophir G. et al.. Human apoE3 but not apoE4 rescues impaired astrocyte activation in apoE null mice. Neurobiology of disease 12, 56–64(2003).
    1. Chen G. Y. & Nunez G. Sterile inflammation: sensing and reacting to damage. Nature reviews. Immunology 10, 826–837, doi: 10.1038/nri2873(2010).
    1. Yu L., Wang L. & Chen S. Endogenous toll-like receptor ligands and their biological significance. Journal of cellular and molecular medicine 14, 2592–2603, doi: 10.1111/j.1582-4934.2010.01127.x(2010).
    1. Said-Sadier N. & Ojcius D. M. Alarmins, inflammasomes and immunity. Biomedical journal 35, 437–449, doi: 10.4103/2319-4170.104408(2012).
    1. Westermark G. T., Westermark P., Berne C., Korsgren O. & Nordic Network for Clinical Islet, T. Widespread amyloid deposition in transplanted human pancreatic islets. The New England journal of medicine 359, 977–979, doi: 10.1056/NEJMc0802893(2008).
    1. Stewart C. R. et al.. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nature immunology 11, 155–161, doi: 10.1038/ni.1836(2010).
    1. Barish G. D. et al.. Bcl-6 and NF-kappaB cistromes mediate opposing regulation of the innate immune response. Genes & development 24, 2760–2765, doi: 10.1101/gad.1998010(2010).
    1. Barish G. D. et al.. The Bcl6-SMRT/NCoR cistrome represses inflammation to attenuate atherosclerosis. Cell metabolism 15, 554–562, doi: 10.1016/j.cmet.2012.02.012(2012).
    1. Basso K. et al.. Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood 115, 975–984, doi: 10.1182/blood-2009-06-227017(2010).
    1. Lei B. et al.. Neuroprotective pentapeptide CN-105 improves functional and histological outcomes in a murine model of intracerebral hemorrhage. Sci Rep 6, 34834, doi: 10.1038/srep34834(2016).
    1. Nishitsuji K., Hosono T., Nakamura T., Bu G. & Michikawa M. Apolipoprotein E regulates the integrity of tight junctions in an isoform-dependent manner in an in vitro blood-brain barrier model. The Journal of biological chemistry 286, 17536–17542, doi: 10.1074/jbc.M111.225532(2011).
    1. Weisgraber K. H. Apolipoprotein E: structure-function relationships. Advances in protein chemistry 45, 249–302(1994).
    1. Marklund N. & Hillered L. Animal modelling of traumatic brain injury in preclinical drug development: where do we go from here ? Br J Pharmacol 164, 1207–1229, doi: 10.1111/j.1476-5381.2010.01163.x(2011).
    1. Petraglia A. L., Dashnaw M. L., Turner R. C. & Bailes J. E. Models of Mild Traumatic Brain Injury: Translation of Physiological and Anatomic Injury. Neurosurgery 75, S34–S49, doi: 10.1227/NEU.0000000000000472(2014).
    1. Wang H. et al.. ApolipoproteinE mimetic peptides improve outcome after focal ischemia. Experimental neurology 241, 67–74, doi: 10.1016/j.expneurol.2012.11.027(2013).
    1. Amor S. & Woodroofe M. N. Innate and adaptive immune responses in neurodegeneration and repair. Immunology 141, 287–291, doi: 10.1111/imm.12134(2014).
    1. Le Thuc O., Blondeau N., Nahon J. L. & Rovere C. The complex contribution of chemokines to neuroinflammation: switching from beneficial to detrimental effects. Annals of the New York Academy of Sciences 1351, 127–140, doi: 10.1111/nyas.12855(2015).
    1. Dawson H. N., Cantillana V., Chen L. & Vitek M. P. The tau N279K exon 10 splicing mutation recapitulates frontotemporal dementia and parkinsonism linked to chromosome 17 tauopathy in a mouse model. The Journal of neuroscience: the official journal of the Society for Neuroscience 27, 9155–9168, doi: 10.1523/JNEUROSCI.5492-06.2007(2007).
    1. Hamm R. J., Pike B. R., O’Dell D. M., Lyeth B. G. & Jenkins L. W. The rotarod test: an evaluation of its effectiveness in assessing motor deficits following traumatic brain injury. Journal of neurotrauma 11, 187–196(1994).

Source: PubMed

3
Subskrybuj