Elsevier

Brain Research

Volume 1746, 1 November 2020, 147000
Brain Research

Research report
The contribution of stem cell factor and granulocyte colony-stimulating factor in reducing neurodegeneration and promoting neurostructure network reorganization after traumatic brain injury

https://doi.org/10.1016/j.brainres.2020.147000Get rights and content

Highlights

  • A severe TBI causes long-term axonal overgrowth in the lesion side brain.

  • SCF + G-CSF treatment prevents the severe TBI-induced axonal overgrowth.

  • The TBI-reduced cortical dendrites next to the TBI cavity are increased by SCF + G-CSF.

  • SCF + G-CSF ameliorates the TBI-induced long-term and widespread neurodegeneration.

  • The TBI-impaired cognitive function is improved by SCF + G-CSF treatment.

Abstract

Traumatic brain injury (TBI) is a major cause of death and disability in young adults worldwide. TBI-induced long-term cognitive deficits represent a growing clinical problem. Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are involved in neuroprotection and neuronal plasticity. However, the knowledge concerning reparative efficacy of SCF + G-CSF treatment in post-acute TBI recovery remains incomplete. This study aims to determine the efficacy of SCF + G-CSF on post-acute TBI recovery in young adult mice. The controlled cortical impact model of TBI was used for inducing a severe damage in the motor cortex of the right hemisphere in 8-week-old male C57BL mice. SCF + G-CSF treatment was initiated 3 weeks after induction of TBI. Severe TBI led to persistent motor functional deficits (Rota-Rod test) and impaired spatial learning function (water maze test). SCF + G-CSF treatment significantly improved the severe TBI-impaired spatial learning function 6 weeks after treatment. TBI also caused significant increases of Fluoro-Jade C positive degenerating neurons in bilateral frontal cortex, striatum and hippocampus, and significant reductions in MAP2+ apical dendrites and overgrowth of SMI312+ axons in peri-TBI cavity frontal cortex and in the ipsilateral hippocampal CA1 at 24 weeks post-TBI. SCF + G-CSF treatment significantly reduced TBI-induced neurodegeneration in the contralateral frontal cortex and hippocampal CA1, increased MAP2+ apical dendrites in the peri-TBI cavity frontal cortex, and prevented TBI-induced axonal overgrowth in both the peri-TBI cavity frontal cortex and ipsilateral hippocampal CA1.These findings reveal a novel pathology of axonal overgrowth after severe TBI and demonstrate a therapeutic potential of SCF + G-CSF in ameliorating severe TBI-induced long-term neuronal pathology, neurostructural network malformation, and impairments in spatial learning.

Introduction

As a growing clinical problem around the world, traumatic brain injury (TBI) remains the leading cause of death and disability in young adults (Maas et al., 2008). The pathological period of post-TBI is divided into 3 phases: an acute phase, a subacute phase, and a chronic phase. The precise duration of the 3 clinical phases is different for individuals because many factors may affect the pathological time course such as their ages and TBI severity. Generally, the acute phase is the first 7 days after TBI, the subacute phase is between 7 days and 3 weeks after TBI, and the chronic phase may begin 3 to 5 weeks after TBI (Edlow et al., 2016, Ge et al., 2018, Missault et al., 2018, Witcher et al., 2018). In severe cases, the chronic phase begins at 3 months post-TBI and continues years after TBI or can even last throughout an individual’s lifespan (Barnes, 1999, Giacino, 2016). To date, the majority of pre-clinical studies have focused on pharmaceutical interventions in neuroprotection in the acute phase of TBI, such as complement inhibition (Rich et al., 2016), immunomodulation (Yen et al., 2018), angiotensin II receptor blockage (Villapol et al., 2015), and cerebral infusion of insulin-like growth factor-1 (Carlson and Saatman, 2018). However, little work has been done in exploring pharmaceutical approaches in post-acute phases of TBI.

The process of the secondary brain injury is progressive, and secondary neuron loss happens in both the ipsilateral and contralateral hemispheres. A recent study has revealed a long-term and progressive neuropathology in bilateral hemispheres up to one year after a single severe brain injury in a mouse model of controlled cortical impact (CCI) (Pischiutta et al., 2018). Moreover, progressive neurodegeneration after CCI-TBI has been found in both the ipsilateral and contralateral hemispheres (Hall et al., 2005, Hall et al., 2008). Widespread neurodegeneration after TBI has been thought to be the results of secondary brain injury and lead to cognitive impairments (Cruz-Haces et al., 2017). In addition to widespread neurodegeneration, reduced functional connectivity in both hemispheres has also been observed in TBI patients (Alhourani et al., 2016). TBI-induced loss of dendrites and axons is also related with cognitive impairments (Rubovitch et al., 2017, Sen et al., 2017). The pharmaceutical intervention for inhibiting widespread neurodegeneration and enhancing neural network reorganization may restrict TBI-induced progressive neuropathology and improve functional outcomes after TBI.

Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are the key hematopoietic growth factors to regulate bone marrow stem cell proliferation, differentiation and mobilization (Greenbaum and Link, 2011, McNiece and Briddell, 1995, Welte et al., 1985, Zsebo et al., 1990). Accumulating evidence shows that SCF and G-CSF also play a role in neuroprotection and neuronal plasticity. Many studies have demonstrated that administration of SCF (Zhao et al., 2007c), G-CSF (Schneider et al., 2005, Sehara et al., 2007, Shyu et al., 2004, Six et al., 2003, Solaroglu et al., 2006, Zhao et al., 2007c), or SCF + G-CSF (Kawada et al., 2006, Toth et al., 2008, Zhao et al., 2007c) leads to reduction in infarction size and amelioration of neurological deficits in experimental stroke. In addition to neuroprotection, SCF has been shown to promote neurite outgrowth (Hirata et al., 1993, Zhao et al., 2007a), and lack of SCF impairs spatial and learning memory (Katafuchi et al., 2000). G-CSF deficient mice also show cognitive problems, impairments in long-term potentiation, and reductions in dendrites in the hippocampus (Diederich et al., 2009). SCF and G-CSF have been demonstrated to cross the blood–brain barrier in intact animals (Zhao et al., 2007b). SCF and G-CSF combined treatment (SCF + G-CSF) has shown synergistic effects in enhancing neurite outgrowth (Su et al., 2013) and neural network reorganization in chronic stroke (Zhao et al., 2007a). We have also demonstrated that SCF + G-CSF treatment in the chronic phase of experimental stroke synergistically improves functional recovery (Zhao et al., 2007a). SCF + G-CSF-improved functional recovery in the chronic phase of experimental stroke is dependent on the SCF + G-CSF-enhanced neurostructural network remodeling (Cui et al., 2015, Cui et al., 2016). Our previous study has demonstrated therapeutic efficacy of SCF + G-CSF in the earlier subacute phase of severe CCI-TBI (Toshkezi et al., 2018a). However, it remains unexplored whether this pharmaceutical approach is effective at the late stage of TBI. The knowledge concerning reparative efficacy of SCF + G-CSF treatment in post-acute TBI remains incomplete.

The aim of the present study is to determine and validate the efficacy of SCF + G-CSF treatment in post-acute TBI on brain repair and functional recovery.

Section snippets

SCF + G-CSF treatment does not improve TBI-impaired cognitive and motor functions 2 weeks after treatment

To ensure that TBI mice presented equal pathological conditions in the treated and non-treated groups before the SCF + G-CSF treatment initiation, we examined cognitive and motor functional deficits one week before treatment by a brief of water maze test and Rota-Rod test (3d test), respectively (Fig. 1). SCF + G-CSF or an equal volume of vehicle solution was subcutaneously injected for 7 consecutive days starting on day 21 post-TBI (Fig. 1). The latency to find the platform was longer in all

Discussion

In the present study, we have demonstrated that the combination treatment of SCF and G-CSF at 3 weeks post-TBI ameliorates severe TBI-induced long-term impairments in spatial learning, reduces neurodegeneration, and enhances neurostructural network reorganization.

SCF + G-CSF treatment improves spatial learning 6 weeks, but not 2 weeks, after treatment in a severe CCI-TBI model. This observation suggests that SCF + G-CSF requires a prolonged period (6 weeks) to repair the brain damaged by severe

Experimental procedure

The animal experiments followed ethical guidelines of the Animal Research, Reporting In Vivo Experiments (ARRIVE). All procedures in this study were approved by the Institutional Animal Care and Use Committee and performed in accordance with the Guide for the Care and Use of Laboratory Animals by the National Institutes of Health.

CRediT authorship contribution statement

JCH contributed to data curation, data analysis, data validation, and writing original draft of manuscript. TR conducted TBI and behavioral tests. XCQ performed brain lesion volume assessment. FH assisted in data analysis. MK performed brain sectioning and histochemical staining. LC contributed to funding acquisition, manuscript review and editing. LRZ contributed to conceptualization, funding acquisition, project administration and supervision, and final revision of the manuscript.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

This study was supported by Merit Review Award (I01RX002125) from the United States Department of Veterans Affairs Rehabilitation Research & Development Service.

References (62)

  • I. Six

    Beneficial effect of pharmacological mobilization of bone marrow in experimental cerebral ischemia

    Eur. J. Pharmacol.

    (2003)
  • I. Solaroglu

    Anti-apoptotic effect of granulocyte-colony stimulating factor after focal cerebral ischemia in the rat

    Neuroscience

    (2006)
  • Z.E. Toth

    The combination of granulocyte colony-stimulating factor and stem cell factor significantly increases the number of bone marrow-derived endothelial cells in brains of mice following cerebral ischemia

    Blood

    (2008)
  • L.R. Zhao

    Hematopoietic growth factors pass through the blood-brain barrier in intact rats

    Exp. Neurol.

    (2007)
  • K.M. Zsebo

    Identification, purification, and biological characterization of hematopoietic stem cell factor from buffalo rat liver–conditioned medium

    Cell

    (1990)
  • A. Alhourani

    Magnetoencephalography-based identification of functional connectivity network disruption following mild traumatic brain injury

    J. Neurophysiol.

    (2016)
  • M.P. Barnes

    Rehabilitation after traumatic brain injury

    Br. Med. Bull.

    (1999)
  • P.S. Buckmaster et al.

    Network properties of the dentate gyrus in epileptic rats with hilar neuron loss and granule cell axon reorganization

    J. Neurophysiol.

    (1997)
  • P.S. Buckmaster et al.

    In vivo intracellular analysis of granule cell axon reorganization in epileptic rats

    J. Neurophysiol.

    (1999)
  • L. Caracciolo

    CREB controls cortical circuit plasticity and functional recovery after stroke

    Nat. Commun.

    (2018)
  • S.W. Carlson et al.

    Central infusion of insulin-like growth factor-1 increases hippocampal neurogenesis and improves neurobehavioral function after traumatic brain injury

    J. Neurotrauma

    (2018)
  • J.F. Carnahan et al.

    Stem cell factor is a neurotrophic factor for neural crest-derived chick sensory neurons

    J. Neurosci.

    (1994)
  • M. Cruz-Haces

    Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases

    Transl. Neurodegener.

    (2017)
  • L. Cui

    Reestablishing neuronal networks in the aged brain by stem cell factor and granulocyte-colony stimulating factor in a mouse model of chronic stroke

    PLoS ONE

    (2013)
  • L. Cui

    Repairing the brain by SCF+G-CSF treatment at 6 months postexperimental stroke: mechanistic determination of the causal link between neurovascular regeneration and motor functional recovery

    ASN Neuro

    (2016)
  • K.M. Dhandapani

    Neuroprotection by stem cell factor in rat cortical neurons involves AKT and NFkappaB

    J. Neurochem.

    (2005)
  • K. Diederich

    The role of granulocyte-colony stimulating factor (G-CSF) in the healthy brain: a characterization of G-CSF-deficient mice

    J. Neurosci.

    (2009)
  • B.L. Edlow

    Diffusion tensor imaging in acute-to-subacute traumatic brain injury: a longitudinal analysis

    BMC Neurol.

    (2016)
  • Giacino, J.T.C., A.; Bergin, M.J.G., 2016. Rehabilitation Access and Outcome After Severe Traumatic Brain Injury: A TBI...
  • A.M. Greenbaum et al.

    Mechanisms of G-CSF-mediated hematopoietic stem and progenitor mobilization

    Leukemia

    (2011)
  • E.D. Hall

    Spatial and temporal characteristics of neurodegeneration after controlled cortical impact in mice: more than a focal brain injury

    J. Neurotrauma

    (2005)
  • View full text