Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Nov 1.
Published in final edited form as: Discov Med. 2010 Nov;10(54):434–442.

Neurorestorative Treatments for Traumatic Brain Injury

Ye Xiong 1, Asim Mahmood 1, Michael Chopp 2,3,*
PMCID: PMC3122155  NIHMSID: NIHMS299898  PMID: 21122475

Abstract

Traumatic brain injury (TBI) remains a major cause of death and permanent disability worldwide, especially in children and young adults. A total of 1.5 million people experience head trauma each year in the United States, with an annual economic cost exceeding $56 billion. Unfortunately, almost all Phase III TBI clinical trials have yet to yield a safe and effective neuroprotective treatment, raising questions regarding the use of neuroprotective strategies as the primary therapy for acute brain injuries. Recent preclinical data suggest that neurorestorative strategies that promote angiogenesis (formation of new blood vessels from pre-existing endothelial cells), axonal remodeling (axonal sprouting and pruning), neurogenesis (generation of new neurons) and synaptogenesis (formation of new synapses) provide promising opportunities for the treatment of TBI. This review discusses select cell-based and pharmacological therapies that activate and amplify these endogenous restorative brain plasticity processes to promote both repair and regeneration of injured brain tissue and functional recovery after TBI.

Keywords: angiogenesis, functional recovery, neurogenesis, plasticity, synaptogenesis, traumatic brain injury

Introduction

Traumatic brain injury (TBI) is a leading cause of mortality and morbidity worldwide, particularly among the young. Neuroprotection is an important strategy for the treatment of TBI (Narayan et al., 2002). To date, no effective neuroprotective agents have been identified from TBI clinical trials. The disappointing clinical trials may be due to variability in treatment approaches and heterogeneity of the population of TBI patients. Another important aspect is that most clinical trial strategies have used drugs that target a single pathophysiological mechanism, although many mechanisms are involved in secondary injury after TBI. Recent research has focused increasingly on multifunctional agents that target multiple injury mechanisms, particularly those that occur later after the insult (Stoica et al., 2009). Targeting multiple injury mechanisms that contribute to the secondary injury cascade may increase successful clinical trial outcomes.

Recent preclinical studies have revealed that TBI induces neurogenesis, axonal sprouting, and angiogenesis (Lu et al., 2004a; Lu et al., 2005; Oshima et al., 2009; Richardson et al., 2007; Xiong et al., 2010a; Zhang et al., 2010), which may contribute to the spontaneous functional recovery. Agents and treatments that promote these neurorestorative processes have been demonstrated to improve functional recovery after brain injury (Zhang and Chopp, 2009). However, clinical trials in TBI have primarily targeted neuroprotection, and trials directed specifically at neurorestoration have not been conducted. The promotion of neurorestorative processes may be a potential therapy for TBI. We review select cell-based and pharmacological therapies that enhance endogenous restorative brain plasticity processes to improve functional recovery after TBI.

Neurogenesis

Throughout life, neurogenesis occurs in all mammalian brains in the subventricular zone (SVZ) of the lateral ventricle and in the dentate gyrus subgranular zone (SGZ) of the hippocampus (Zhao et al., 2008). Newly generated neurons originate from neural stem cells (NSCs) in the adult brain. NSCs are self-renewing multipotent cells that generate glial and neuronal cells (Zhao et al., 2008). Granule neurons in the dentate gyrus of the hippocampus continuously die, and the neural stem/progenitor cells in the SGZ may proliferate to maintain a constant cell number in the dentate gyrus. Moreover, newly generated neurons in the dentate gyrus are capable of projecting axons into the CA3 region of the hippocampus in normal brains in rodents (Hastings and Gould, 1999). TBI induces hippocampal cell proliferation (Kernie et al., 2001; Lu et al., 2005; Xiong et al., 2008), and the vast majority of the newly generated cells in the SGZ that survive for 10 weeks after TBI differentiate into mature neurons (Sun et al., 2007). Newborn neurons extend axonal projections into the CA3 region as early as 2 weeks after TBI (Emery et al., 2005), which may contribute to cognitive recovery observed in rats that have experienced a TBI. In the normal adult brain, SVZ-derived neuroblasts migrate along the rostral migratory stream to the olfactory bulb, where these cells differentiate into interneurons to replace those that have died. After cortical injuries, a portion of neuroblasts generated in the SVZ migrate to injured areas instead of the rostral migratory stream. Following TBI, neuroblasts migrating from the SVZ can differentiate into neurons and glia (Kernie et al., 2001).

Angiogenesis

The adult central nervous system (CNS) vasculature is extremely stable under physiological conditions, but is activated after injury (Greenberg and Jin, 2005). Adult vascular remodeling includes angiogenesis by mature endothelial cells (that is, the formation of new capillaries from pre-existing vessels) and vasculogenesis (de novo formation of blood vessels when there are no pre-existing ones) by endothelial progenitor cells (EPCs). EPCs are present in the bone marrow and peripheral blood, and mobilize to the latter following TBI (Guo et al., 2009). There is a substantial increase in vasculogenesis following TBI (Morgan et al., 2007). Pharmacological agents such as erythropoietin (EPO) and statins increase the number, mobilization and functional activity of EPCs (Besler et al., 2008). EPO, statins, bone marrow stromal cells (MSCs), and thymosin beta4 promote angiogenesis and improve functional recovery in rats after TBI (Chopp and Li, 2002; Lu et al., 2004b; Lu et al., 2007b; Mammis et al., 2009; Wible and Laskowitz, 2010; Xiong et al., 2010a; Xiong et al., 2010b). The strategies for mobilization and/or transplantation of EPCs or treatment with angiogenesis-enhancing agents may emerge as promising approaches for the treatment of TBI.

Coupling of Neurogenesis and Angiogenesis

Neurovascular niches within the CNS consist of endothelial cells, pericytes, neurons and glial cells, as well as growth factors and extracellular matrix proteins surrounding the endothelium (Lok et al., 2007). The neurovascular niches provide microenvironments for NSCs in the adult brain; newly generated, immature neurons are closely associated with the remodeling vasculature. The generation of new vasculature facilitates coupled neurorestorative processes including neurogenesis and synaptogenesis, which improve functional recovery (Li and Chopp, 2009; Zhang and Chopp, 2009). Angiogenesis and neurogenesis may play a significant role in mediating functional recovery following experimental TBI (Chopp et al., 2008; Li and Chopp, 2009; Lu et al., 2005; Wu et al., 2008b; Xiong et al., 2008; Zhang et al., 2009b). Neurorestorative agents that increase angiogenesis and neurogenesis have been shown to improve functional outcome following brain injury (Zhang et al., 2009b; Zhang and Chopp, 2009). Vascular endothelial cells within the neurovascular niche affect neurogenesis directly via contact with neural progenitor cells while soluble factors from the vascular system that are released into the CNS enhance neurogenesis via paracrine signaling (Yang et al., 2010). A better understanding of precise molecular mechanisms in the neurovascular niches will be important for developing novel angiogenic and neurogenic therapies for brain injuries.

Axonal Remodeling

The CNS has a limited capacity to regenerate after injury. Axonal sprouting from surviving neurons may be associated with spontaneous motor recovery over time after TBI (Oshima et al., 2009; Smith et al., 2007). Spontaneous pericontusional axon sprouting takes place within 1–2 weeks after TBI, which is induced by controlled cortical impact (CCI) in the adult rat but ultimately fails due to an axonal growth-inhibitory environment (Harris et al., 2010). To reduce pericontusional growth-inhibitory chondroitin sulphate proteoglycans, acute infusion of chondroitinase ABC into the site of the cortical contusion was performed, which enhanced and prolonged the sprouting response and reduced unskilled limb use deficits (Harris et al., 2010). In principle, a treatment that promotes axonal plasticity could be beneficial to functional recovery after brain injury (Smith et al., 2007). The corticospinal tract is a major fiber bundle arising from layer V pyramidal neurons of the frontal motor cortex and connects via the corticospinal or pyramidal tracts to contralateral motor neurons of the spinal cord to control voluntary movements. Collateral sprouting of the unlesioned corticospinal tract at the cervical spinal cord and neuromotor functional recovery were observed following unilateral TBI in mice (Oshima et al., 2009). In a full-thickness lesion of the forelimb region of the sensorimotor cortex, skilled paw-reaching behavior, a task that requires corticospinal function, was only partially recovered by 4 weeks. Inosine infused into the lateral ventricles for 4 weeks produced an almost complete recovery of skilled paw-reaching ability, which is associated with sprouting of the uninjured corticospinal axons across the midline into the red nucleus and cervical cord of the lesioned pathway (Smith et al., 2007).

Synaptogenesis

Brain function relies on communication among neurons through highly specialized contacts (that is, the synapses) and synaptic dysfunction plays a critical role in injury-induced defects of the CNS. In response to a CNS injury, surviving neurons reorganize their connections and form new synapses to replace those lost caused by the lesion (Becher et al., 1999). Synaptophysin (SYP), a neuronal marker of synaptogenesis, is an integral transmembrane protein component of presynaptic vesicles and is widely expressed in neurons. CCI results in the loss of specific neurons in the CA3 subfield of the ipsilateral hippocampus, resulting in partial loss of afferents to the CA1 subfield; the CNS compensates for deafferentiation by initiating synaptogenesis capable of restoring some of the lost synaptic contacts (Scheff et al., 2005). After CCI, the density of SYP signals in the injury boundary zone was less than that of the intact cortical area (Lu et al., 2004a). After treatment with atorvastatin, the density significantly increased in this area compared to the control rats (Lu et al., 2004a). Atorvastatin may protect synapses from the impact or induce synaptogenesis in the boundary zone. Almost no SYP-positive signals were detected in the stratum lucidum and some weak signals were observed in the pyramidal cell layer. After atorvastatin treatment, intense SYP signals were found in the pyramidal cell layer as well as in the stratum lucidum (Lu et al., 2004a). Atorvastatin-induced synaptogenesis may contribute to reduction in the neurological functional deficits.

Bone Marrow Stromal Cells

Although human embryonic stem cells (hESCs) or fetal tissues are suitable sources for cell-based therapies, their clinical application is limited by both ethical considerations and other practical challenges including tumorigenicity, cell viability and antigenic compatibility. Reprogramming differentiated cells generates induced pluripotent stem cells (iPSCs) that resemble embryonic stem cells (Yamanaka, 2007). These iPSCs avoid the ethical issues and remove the major roadblock of immune rejection associated with the clinical use of hESCs, as well as potentially generate patient-specific cells for cell-replacement therapy. However, the safety and therapeutic applications of iPSCs and iPSC-derived cells must be rigorously tested in appropriate animal models before advancing to any clinical trial. The most important issue with iPSCs is potential tumorigenicity. Even with improvements in the virus-free and transgene-free reprogramming technologies, the cancer-causing possibility of the derived “safe” iPSCs/derivatives still needs to be evaluated in animal models before their clinical application for regenerative treatment.

Bone marrow stromal cells (MSCs) are a mixed cell population, including stem and progenitor cells, and are a promising source of cell-based therapy for TBI, since they can be easily isolated from many tissues and expanded in culture from patients without ethical and immune rejection problems (Chopp and Li, 2002). When grafted into the lateral ventricles of neonatal mouse brains, mouse MSCs migrated and differentiated into olfactory bulb granule cells and periventricular astrocytes (Deng et al., 2006). Systematically infused rat MSCs migrated into injured rat brains and survived (Lu et al., 2001). Some of the implanted MSCs expressed cell markers for neurons and astrocytes. Expression of the chemokine stromal-cell-derived factor-1 was significantly increased in the lesion boundary zone after brain injury induced by ischemia (Shen et al., 2007). The stromal-cell-derived factor-1 receptor, CXC-chemokine receptor-4, was expressed in MSCs both in vitro and in vivo (Shen et al., 2007). The interaction of stromal-cell-derived factor-1 with CXC-chemokine receptor-4 may contribute to the trafficking of transplanted MSCs into the injured brain (Itoh et al., 2009; Shen et al., 2007). Direct implantation (6 h post injury) of MSCs enhances neuroprotection via activation of resident NSC nuclear factor κB activity leading to an increase in interleukin-6 production and decrease in apoptosis (Walker et al., 2010). The delayed administration (24 h or 1 week following injury) of MSCs also significantly improved functional outcome in rodents following TBI (Chopp and Li, 2002; Chopp et al., 2009; Lu et al., 2001; Mahmood et al., 2004b; Mahmood et al., 2005; Mahmood et al., 2006).

MSCs secrete various growth factors, including brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF) and bFGF (basic fibroblast growth factor), and increase the levels of these factors in the brain (Chopp and Li, 2002; Mahmood et al., 2004a). MSCs also induce intrinsic parenchymal cells to produce these growth factors (Mahmood et al., 2004a). After MSC transplantation, these neurotrophic/growth factors enhance angiogenesis and vascular stabilization in the lesion boundary zone where the majority of MSCs that survive in the brain are located (Mahmood et al., 2006). These growth factors also promote neurogenesis in vitro and in vivo (Jin et al., 2002; Lee et al., 2002; Yoshimura et al., 2003). In rodent TBI models, MSCs not only increased vascular density in the lesion boundary zone and hippocampus (Qu et al., 2008), but also enhanced neurogenesis in the SGZ and SVZ (Mahmood et al., 2004b). Delayed (4 days after TBI) treatment with MSCs alone did not reduce lesion volume, whereas MSCs seeded in collagen scaffolds significantly reduced lesion volume, enhanced the migration of MSCs into the lesion boundary zone, and significantly improved spatial learning and sensorimotor function (Lu et al., 2007a). Even more delayed (7 days post injury) transplantation of MSCs or MSCs seeded in scaffolds improved spatial learning and sensorimotor function, enhanced angiogenesis in the injured cortex and the ipsilateral hippocampus and increased transcallosal neural fibers in the injured cortex (Xiong et al., 2009). The significant therapeutic benefits of MSCs are not attributed to the few MSCs that differentiate into neural cells (Lu et al., 2001). However, MSCs appear to act as neurotrophic/growth factor generators and inducers to promote brain functional recovery via angiogenesis, neurogenesis, synaptogenesis and axonal remodeling (Chopp and Li, 2002; Chopp and Li, 2006). MSCs (or neural stem/precursor cells)-seeded scaffolds may be a new and effective strategy for treatment of TBI.

The safety and feasibility of treatment with autologous MSCs were assessed in seven patients with TBI (Zhang et al., 2008). In this trial, no toxicity related to the cell therapy was observed within the 6-month follow-up period. A safety study of autologous stem cell treatment in children with TBI has also been completed (ClinicalTrials.gov, Identifier: NCT00254722); however, no data are available. This study should determine if bone marrow harvest and re-infusion is safe in children after severe TBI.

Erythropoietin

EPO stimulates the maturation, differentiation and survival of hematopoietic progenitor cells to maintain erythropoiesis and has been widely used for treatment of anemia. Although low levels of EPO and EPO receptors exist in normal adult brains, increased expression of EPO and the EPO receptors is found in neurons, neural progenitor cells, glial cells, and endothelial cells in response to injury (Grasso et al., 2004). EPO (5000 U/kg ip) was demonstrated to cross the blood-brain barrier and to protect against brain injury in rats (Brines et al., 2000; Wang et al., 2004). Acute EPO administration (within 6-h post-TBI) provides neuroprotection (that is, decreased lesion volume and cell loss) as well as enhances neurogenesis, and subsequently improves sensorimotor and spatial learning functions in rat and mouse models (Brines et al., 2000; Cherian et al., 2007; Xiong et al., 2008; Zhang et al., 2009b). Delayed administration of EPO (5000 U/kg, ip for 14 days) from day 1 following TBI in rats significantly increased dentate gyrus neurogenesis and improved spatial memory (Lu et al., 2005). Post-TBI treatment (6-h or 24-h post-injury) with EPO (5000 U/kg) significantly increased the expression of BDNF and improved spatial learning following injury in rats (Mahmood et al., 2007b). Our recent studies demonstrate that a multiple-dose treatment with EPO (5000 U/kg/day for 3 days initiated at day 1 post-injury) is more effective than a single-dose EPO therapy in improving functional recovery in rats after TBI (Xiong et al., 2010a). Treatment with EPO also contributes to neurovascular remodeling, leading to improved neurobehavioral outcomes following TBI (Xiong et al., 2010a; Zhang et al., 2009b). EPO enhances VEGF secretion from neural progenitor cells; the treatment of such cells with EPO leads to the upregulation of VEGF receptor 2 expression in cerebral endothelial cells, promoting angiogenesis (Wang et al., 2008).

Our previous study showed that delayed (24 h post injury) EPO treatment improves neurological functional recovery without reducing lesion volume after TBI (Xiong et al., 2010a). In addition to its effects on neurogenesis and angiogenesis, EPO may improve neurological recovery partially through enhancement of axonal plasticity. Axonal sprouting from the intact corticospinal tract was increased in the denervated side of the gray matter of both cervical and lumbar levels of the spinal cord at day 35 after TBI. However, the corticospinal tract axonal sprouting was significantly enhanced at both cervical and lumbar spinal cord in the EPO-treated TBI animals (Zhang et al., 2010). The contralesional corticospinal tract axonal sprouting was highly and positively correlated with sensorimotor recovery after TBI, suggesting axonal sprouting induced by EPO treatment may contribute to functional recovery after TBI.

In a small clinical trial for treating stroke with EPO, intravenous administration of EPO is well tolerated in acute ischemic stroke and associated with an improvement in clinical outcome at one month (Ehrenreich et al., 2002). However, in a recent large clinical trial of stroke, EPO treatment has not resulted in benefits (increasing higher mortality compared to placebo controls) (Ehrenreich et al., 2009). Combination of tissue plasminogen activator with EPO may be one of the important factors for this failed clinical trial, because a very large number of EPO-treated stroke patients also received tissue plasminogen activator treatment and combination of EPO with tissue plasminogen activator has been demonstrated to cause detrimental effects in animal models of stroke (Jia et al., 2010).

A phase III trial of EPO in patients with TBI has been planned (ClinicalTrials.gov, NCT00987454). A phase II trial investigating the safety of treatment with Darbepoetin Alfa (a long-acting form of EPO) in patients with severe TBI is ongoing (ClinicalTrials.gov, NCT00375869). A phase II/III trial to investigate the early administration of EPO to TBI patients is ongoing (ClinicalTrials.gov, NCT00260052). The high doses of EPO used for the treatment of stroke and TBI significantly increased hematocrit (Mahmood et al., 2007b; Xiong et al., 2008), which may cause adverse vascular effects such as deep venous thrombosis (Lapchak, 2008). However, non-hematopoietic EPO analogs, such as the carbamylated form of EPO (CEPO), are as effective as hematopoietic EPO in neuroprotection and are not associated with the hematopoietic side effects (Lapchak, 2008; Mahmood et al., 2007b), indicating their potential application to TBI therapy. The optimal EPO dose, dosing interval, and number of doses for reducing brain injury that promote neurorestoration and improve functional recovery have not been fully investigated after TBI. The EPO doses for the TBI clinical trials are based on stroke trials. Lack of preclinical data on these important aspects highlights the importance of fully evaluating EPO and its analogs for both acute protection and chronic restoration of function after TBI.

Statins

Statins, inhibitors of cholesterol biosynthesis used to lower cholesterol levels, induce angiogenesis, neurogenesis and synaptogenesis, and enhance functional recovery following TBI in rats (Lu et al., 2004a; Lu et al., 2004b; Lu et al., 2007b; Wu et al., 2008b). These beneficial effects of statins are independent of cholesterol-lowering action. Beneficial effects of simvastatin may be mediated through activation of Akt, Forkhead transcription factor 1 and nuclear factor–κB signaling pathways, which suppress the activation of caspase-3 and apoptotic cell death, and thereby, lead to neuronal function recovery after TBI (Wu et al., 2008a). Simvastatin activates the Akt-mediated signaling pathway, subsequently upregulating the expression of growth factors and inducing neurogenesis in the dentate gyrus of the hippocampus, thereby leading to restoration of cognitive function after TBI in rats (Wu et al., 2008b). In addition, simvastatin treatment provided long-lasting (3 month) functional improvement following TBI in rats (Mahmood et al., 2009). The protective mechanisms of statins may be partly attributed to a reduction in the inflammatory response following TBI (Li et al., 2009). When administered in combination with MSCs in a rat model of TBI, atorvastatin increased MSC access and/or survival within the injured brain and enhanced functional recovery compared with either MSC or atorvastatin monotherapy (Mahmood et al., 2007a), suggesting that statins might be used in conjunction with MSC transplantation for treating neurological disorders and injuries.

Given the wide use, favorable safety profile and positive clinical data for statins, the rare occurrence of serious adverse events and the extensive available preclinical data demonstrating neuroprotection and neurorestoration (Wible and Laskowitz, 2010), further clinical trials are warranted to determine the neuroprotective and neurorestorative properties of statins following TBI. The effect of rosuvastatin on TBI-induced cytokine change is ongoing in a phase I/II trial (ClinicalTrials.gov, NCT00990028).

Thymosin Beta 4

Thymosin beta 4 (Tβ4), a polypeptide of 43-amino acids, was first isolated from bovine thymus tissue and subsequently found to exist in all mammals studied. The major intracellular function of Tβ4 is G-actin-sequestration, which is necessary for cell motility and organogenesis (Crockford, 2007). Recent studies demonstrate that Tβ4 is a multifunctional peptide. It inhibits inflammation and apoptosis, and promotes tissue repair in skin, cornea, and heart (Morris et al., 2010b). Tβ4 is an essential paracrine factor of EPCs, and Tβ4 promotes angiogenesis after ischemic injury (Smart et al., 2007). Safety, tolerability and efficiency of Tβ4 are being evaluated in clinical patients with acute myocardial infarction (Crockford, 2007).

Tβ4 plays a critical role in many cellular processes including mobility, axonal path-finding, neurite formation, proliferation and neuronal survival (Morris et al., 2010b; Sun and Kim, 2007). Our recent study demonstrates that Tβ4 improves neurological functional recovery in mice with experimental autoimmune encephalomyelitis (Zhang et al., 2009a) and in rats with embolic stroke (Morris et al., 2010a). Tβ4 is a potential treatment for TBI. Tβ4 (6 mg/kg) was administered ip starting at day 1 and then every 3 days for an additional 4 doses to the TBI rats (Xiong et al., 2010b). Neurological functional recovery was evaluated. Animals were euthanized 35 days after injury and brain sections were stained for immunohistochemistry to assess angiogenesis, neurogenesis, and oligodendrogenesis after Tβ4 treatment. Delayed Tβ4 treatment did not affect lesion volume but significantly reduced hippocampal cell loss, enhanced angiogenesis and neurogenesis in the injured cortex and hippocampus, increased oligodendrogenesis in the CA3 region, and significantly improved sensorimotor functional recovery and spatial learning compared to the saline treatment. These data demonstrate that administration of Tβ4 significantly improves histological and functional outcomes in rats with TBI, indicating that Tβ4 has considerable therapeutic potential in TBI patients. Further investigation of Tβ4 is warranted for the treatment of TBI.

Conclusion

TBI induces angiogenesis, axonal remodeling, and neurogenesis in preclinical studies. Strategies that enhance these neurorestorative processes have been demonstrated to improve brain functional recovery in experimental TBI. A better understanding of the relationship between functional recovery and these processes will lead to novel therapeutic strategies for the treatment of TBI. The cell-based and pharmacological therapies (for example, MSCs, EPO, CEPO, statins, Tβ4, alone or in combination) described in this review induce endogenous neurorestorative processes by increasing angiogenesis, axonal remodeling, neurogenesis and synaptogenesis, and consequently improve neurological functional recovery following TBI. However, several issues should be considered during the preclinical studies and clinical trials of these strategies in TBI. Prior to the translation of an agent or cell therapy into TBI clinical trials, sufficient preclinical data should be obtained from multiple experiments, preferably in several brain injury models, on optimal administration routes, single dose versus multiple dose, bolus dose versus continuous infusion, dose-response, and therapeutic windows. Extensive pharmacokinetic data for agents to treat injured brains should also be obtained, ensuring an adequate concentration in the brain tissue. In addition, the effective progression of strategies into clinical trials may require multiple functional agents including EPO, CEPO, statins, Tβ4, or combination therapies. These potential combinations include single agents (for example, small molecules or cytokines including EPO, CEPO, Tβ4, VEGF) with cells (for example, MSCs, NSCs, iPSCs and genetically modified derivatives) or with other approaches (biomaterial scaffolds, physical or electrical stimulation). For the safety and efficacy, the interaction of agents used in combination therapy (such as EPO combined with tissue plasminogen activator) should be fully addressed in preclinical studies before their translation to clinical trials. Although it is still important to further investigate neuroprotective treatments for TBI, an interesting novel research direction is the development of neurorestorative strategies that enhance axonal remodeling, angiogenesis, neurogenesis and synaptogenesis to improve functional recovery of the injured brain.

Acknowledgments

The authors’ research was supported by NINDS grants RO1 NS62002 and PO1 NS042259.

References

  1. Becher A, Drenckhahn A, Pahner I, Margittai M, Jahn R, Ahnert-Hilger G. The synaptophysin-synaptobrevin complex: a hallmark of synaptic vesicle maturation. J Neurosci. 1999;19(6):1922–1931. doi: 10.1523/JNEUROSCI.19-06-01922.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Besler C, Doerries C, Giannotti G, Luscher TF, Landmesser U. Pharmacological approaches to improve endothelial repair mechanisms. Expert Rev Cardiovasc Ther. 2008;6(8):1071–1082. doi: 10.1586/14779072.6.8.1071. [DOI] [PubMed] [Google Scholar]
  3. Brines ML, Ghezzi P, Keenan S, Agnello D, De Lanerolle NC, Cerami C, Itri LM, Cerami A. Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proc Natl Acad Sci U S A. 2000;97(19):10526–10531. doi: 10.1073/pnas.97.19.10526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Cherian L, Goodman JC, Robertson C. Neuroprotection with erythropoietin administration following controlled cortical impact injury in rats. J Pharmacol Exp Ther. 2007;322(2):789–794. doi: 10.1124/jpet.107.119628. [DOI] [PubMed] [Google Scholar]
  5. Chopp M, Li Y. Treatment of neural injury with marrow stromal cells. Lancet Neurol. 2002;1(2):92–100. doi: 10.1016/s1474-4422(02)00040-6. [DOI] [PubMed] [Google Scholar]
  6. Chopp M, Li Y. Transplantation of bone marrow stromal cells for treatment of central nervous system diseases. Adv Exp Med Biol. 2006;585:49–64. doi: 10.1007/978-0-387-34133-0_4. [DOI] [PubMed] [Google Scholar]
  7. Chopp M, Li Y, Zhang J. Plasticity and remodeling of brain. J Neurol Sci. 2008;265(1–2):97–101. doi: 10.1016/j.jns.2007.06.013. [DOI] [PubMed] [Google Scholar]
  8. Chopp M, Li Y, Zhang ZG. Mechanisms underlying improved recovery of neurological function after stroke in the rodent after treatment with neurorestorative cell-based therapies. Stroke. 2009;40(3 Suppl):S143–145. doi: 10.1161/STROKEAHA.108.533141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Crockford D. Development of thymosin beta4 for treatment of patients with ischemic heart disease. Ann N Y Acad Sci. 2007;1112:385–395. doi: 10.1196/annals.1415.051. [DOI] [PubMed] [Google Scholar]
  10. Deng J, Petersen BE, Steindler DA, Jorgensen ML, Laywell ED. Mesenchymal stem cells spontaneously express neural proteins in culture and are neurogenic after transplantation. Stem Cells. 2006;24(4):1054–1064. doi: 10.1634/stemcells.2005-0370. [DOI] [PubMed] [Google Scholar]
  11. Ehrenreich H, Hasselblatt M, Dembowski C, Cepek L, Lewczuk P, Stiefel M, Rustenbeck HH, Breiter N, Jacob S, Knerlich F, Bohn M, Poser W, Ruther E, Kochen M, Gefeller O, Gleiter C, Wessel TC, De Ryck M, Itri L, Prange H, et al. Erythropoietin therapy for acute stroke is both safe and beneficial. Mol Med. 2002;8(8):495–505. [PMC free article] [PubMed] [Google Scholar]
  12. Ehrenreich H, Weissenborn K, Prange H, Schneider D, Weimar C, Wartenberg K, Schellinger PD, Bohn M, Becker H, Wegrzyn M, Jahnig P, Herrmann M, Knauth M, Bahr M, Heide W, Wagner A, Schwab S, Reichmann H, Schwendemann G, Dengler R, et al. Recombinant human erythropoietin in the treatment of acute ischemic stroke. Stroke. 2009;40(12):e647–656. doi: 10.1161/STROKEAHA.109.564872. [DOI] [PubMed] [Google Scholar]
  13. Emery DL, Fulp CT, Saatman KE, Schutz C, Neugebauer E, Mcintosh TK. Newly born granule cells in the dentate gyrus rapidly extend axons into the hippocampal CA3 region following experimental brain injury. J Neurotrauma. 2005;22(9):978–988. doi: 10.1089/neu.2005.22.978. [DOI] [PubMed] [Google Scholar]
  14. Grasso G, Sfacteria A, Cerami A, Brines M. Erythropoietin as a tissue-protective cytokine in brain injury: what do we know and where do we go? Neuroscientist. 2004;10(2):93–98. doi: 10.1177/1073858403259187. [DOI] [PubMed] [Google Scholar]
  15. Greenberg DA, Jin K. From angiogenesis to neuropathology. Nature. 2005;438(7070):954–959. doi: 10.1038/nature04481. [DOI] [PubMed] [Google Scholar]
  16. Guo X, Liu L, Zhang M, Bergeron A, Cui Z, Dong JF, Zhang J. Correlation of CD34+ cells with tissue angiogenesis after traumatic brain injury in a rat model. J Neurotrauma. 2009;26(8):1337–1344. doi: 10.1089/neu.2008.0733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Harris N, Mironova YA, Hovda D, Sutton RL. Chondroitinase ABC enhances pericontusion axonal sprouting but does not confer robust improvements in behavioral recovery. J Neurotrauma. 2010 doi: 10.1089/neu.2010.1470. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Hastings NB, Gould E. Rapid extension of axons into the CA3 region by adult-generated granule cells. J Comp Neurol. 1999;413(1):146–154. doi: 10.1002/(sici)1096-9861(19991011)413:1<146::aid-cne10>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  19. Itoh T, Satou T, Ishida H, Nishida S, Tsubaki M, Hashimoto S, Ito H. The relationship between SDF-1alpha/CXCR4 and neural stem cells appearing in damaged area after traumatic brain injury in rats. Neurol Res. 2009;31(1):90–102. doi: 10.1179/174313208X332995. [DOI] [PubMed] [Google Scholar]
  20. Jia L, Chopp M, Zhang L, Lu M, Zhang Z. Erythropoietin in combination of tissue plasminogen activator exacerbates brain hemorrhage when treatment is initiated 6 hours after stroke. Stroke. 2010;41(9):2071–2076. doi: 10.1161/STROKEAHA.110.586198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Jin K, Zhu Y, Sun Y, Mao XO, Xie L, Greenberg DA. Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci U S A. 2002;99(18):11946–11950. doi: 10.1073/pnas.182296499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Kernie SG, Erwin TM, Parada LF. Brain remodeling due to neuronal and astrocytic proliferation after controlled cortical injury in mice. J Neurosci Res. 2001;66(3):317–326. doi: 10.1002/jnr.10013. [DOI] [PubMed] [Google Scholar]
  23. Lapchak PA. Carbamylated erythropoietin to treat neuronal injury: new development strategies. Expert Opin Investig Drugs. 2008;17(8):1175–1186. doi: 10.1517/13543784.17.8.1175. [DOI] [PubMed] [Google Scholar]
  24. Lee J, Duan W, Mattson MP. Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. J Neurochem. 2002;82(6):1367–1375. doi: 10.1046/j.1471-4159.2002.01085.x. [DOI] [PubMed] [Google Scholar]
  25. Li B, Mahmood A, Lu D, Wu H, Xiong Y, Qu C, Chopp M. Simvastatin attenuates microglial cells and astrocyte activation and decreases interleukin-1beta level after traumatic brain injury. Neurosurgery. 2009;65(1):179–185. doi: 10.1227/01.NEU.0000346272.76537.DC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Li Y, Chopp M. Marrow stromal cell transplantation in stroke and traumatic brain injury. Neurosci Lett. 2009;456(3):120–123. doi: 10.1016/j.neulet.2008.03.096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Lok J, Gupta P, Guo S, Kim WJ, Whalen MJ, Van Leyen K, Lo EH. Cell-cell signaling in the neurovascular unit. Neurochem Res. 2007;32(12):2032–2045. doi: 10.1007/s11064-007-9342-9. [DOI] [PubMed] [Google Scholar]
  28. Lu D, Goussev A, Chen J, Pannu P, Li Y, Mahmood A, Chopp M. Atorvastatin reduces neurological deficit and increases synaptogenesis, angiogenesis, and neuronal survival in rats subjected to traumatic brain injury. J Neurotrauma. 2004a;21(1):21–32. doi: 10.1089/089771504772695913. [DOI] [PubMed] [Google Scholar]
  29. Lu D, Mahmood A, Goussev A, Schallert T, Qu C, Zhang ZG, Li Y, Lu M, Chopp M. Atorvastatin reduction of intravascular thrombosis, increase in cerebral microvascular patency and integrity, and enhancement of spatial learning in rats subjected to traumatic brain injury. J Neurosurg. 2004b;101(5):813–821. doi: 10.3171/jns.2004.101.5.0813. [DOI] [PubMed] [Google Scholar]
  30. Lu D, Mahmood A, Qu C, Goussev A, Schallert T, Chopp M. Erythropoietin enhances neurogenesis and restores spatial memory in rats after traumatic brain injury. J Neurotrauma. 2005;22(9):1011–1017. doi: 10.1089/neu.2005.22.1011. [DOI] [PubMed] [Google Scholar]
  31. Lu D, Mahmood A, Qu C, Hong X, Kaplan D, Chopp M. Collagen scaffolds populated with human marrow stromal cells reduce lesion volume and improve functional outcome after traumatic brain injury. Neurosurgery. 2007a;61(3):596–602. doi: 10.1227/01.NEU.0000290908.38438.B2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Lu D, Mahmood A, Wang L, Li Y, Lu M, Chopp M. Adult bone marrow stromal cells administered intravenously to rats after traumatic brain injury migrate into brain and improve neurological outcome. Neuroreport. 2001;12(3):559–563. doi: 10.1097/00001756-200103050-00025. [DOI] [PubMed] [Google Scholar]
  33. Lu D, Qu C, Goussev A, Jiang H, Lu C, Schallert T, Mahmood A, Chen J, Li Y, Chopp M. Statins increase neurogenesis in the dentate gyrus, reduce delayed neuronal death in the hippocampal CA3 region, and improve spatial learning in rat after traumatic brain injury. J Neurotrauma. 2007b;24(7):1132–1146. doi: 10.1089/neu.2007.0288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Mahmood A, Goussev A, Kazmi H, Qu C, Lu D, Chopp M. Long-term benefits after treatment of traumatic brain injury with simvastatin in rats. Neurosurgery. 2009;65(1):187–191. doi: 10.1227/01.NEU.0000343540.24780.D6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Mahmood A, Lu D, Chopp M. Intravenous administration of marrow stromal cells (MSCs) increases the expression of growth factors in rat brain after traumatic brain injury. J Neurotrauma. 2004a;21(1):33–39. doi: 10.1089/089771504772695922. [DOI] [PubMed] [Google Scholar]
  36. Mahmood A, Lu D, Chopp M. Marrow stromal cell transplantation after traumatic brain injury promotes cellular proliferation within the brain. Neurosurgery. 2004b;55(5):1185–1193. doi: 10.1227/01.neu.0000141042.14476.3c. [DOI] [PubMed] [Google Scholar]
  37. Mahmood A, Lu D, Qu C, Goussev A, Chopp M. Human marrow stromal cell treatment provides long-lasting benefit after traumatic brain injury in rats. Neurosurgery. 2005;57(5):1026–1031. doi: 10.1227/01.neu.0000181369.76323.50. discussion 1026–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Mahmood A, Lu D, Qu C, Goussev A, Chopp M. Long-term recovery after bone marrow stromal cell treatment of traumatic brain injury in rats. J Neurosurg. 2006;104(2):272–277. doi: 10.3171/jns.2006.104.2.272. [DOI] [PubMed] [Google Scholar]
  39. Mahmood A, Lu D, Qu C, Goussev A, Chopp M. Treatment of traumatic brain injury with a combination therapy of marrow stromal cells and atorvastatin in rats. Neurosurgery. 2007a;60(3):546–553. doi: 10.1227/01.NEU.0000255346.25959.99. [DOI] [PubMed] [Google Scholar]
  40. Mahmood A, Lu D, Qu C, Goussev A, Zhang ZG, Lu C, Chopp M. Treatment of traumatic brain injury in rats with erythropoietin and carbamylated erythropoietin. J Neurosurg. 2007b;107(2):392–397. doi: 10.3171/JNS-07/08/0392. [DOI] [PubMed] [Google Scholar]
  41. Mammis A, Mcintosh TK, Maniker AH. Erythropoietin as a neuroprotective agent in traumatic brain injury Review. Surg Neurol. 2009;71(5):527–531. doi: 10.1016/j.surneu.2008.02.040. discussion 531. [DOI] [PubMed] [Google Scholar]
  42. Morgan R, Kreipke CW, Roberts G, Bagchi M, Rafols JA. Neovascularization following traumatic brain injury: possible evidence for both angiogenesis and vasculogenesis. Neurol Res. 2007;29(4):375–381. doi: 10.1179/016164107X204693. [DOI] [PubMed] [Google Scholar]
  43. Morris DC, Chopp M, Zhang L, Lu M, Zhang ZG. Thymosin beta4 improves functional neurological outcome in a rat model of embolic stroke. Neuroscience. 2010a;169(2):674–682. doi: 10.1016/j.neuroscience.2010.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Morris DC, Chopp M, Zhang L, Zhang ZG. Thymosin beta4: a candidate for treatment of stroke? Ann N Y Acad Sci. 2010b;1194:112–117. doi: 10.1111/j.1749-6632.2010.05469.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Narayan RK, Michel ME, Ansell B, Baethmann A, Biegon A, Bracken MB, Bullock MR, Choi SC, Clifton GL, Contant CF, Coplin WM, Dietrich WD, Ghajar J, Grady SM, Grossman RG, Hall ED, Heetderks W, Hovda DA, Jallo J, Katz RL, et al. Clinical trials in head injury. J Neurotrauma. 2002;19(5):503–557. doi: 10.1089/089771502753754037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Oshima T, Lee S, Sato A, Oda S, Hirasawa H, Yamashita T. TNF-alpha contributes to axonal sprouting and functional recovery following traumatic brain injury. Brain Res. 2009;1290:102–110. doi: 10.1016/j.brainres.2009.07.022. [DOI] [PubMed] [Google Scholar]
  47. Qu C, Mahmood A, Lu D, Goussev A, Xiong Y, Chopp M. Treatment of traumatic brain injury in mice with marrow stromal cells. Brain Res. 2008;1208:234–239. doi: 10.1016/j.brainres.2008.02.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Richardson RM, Sun D, Bullock MR. Neurogenesis after traumatic brain injury. Neurosurg Clin N Am. 2007;18(1):169–181. xi. doi: 10.1016/j.nec.2006.10.007. [DOI] [PubMed] [Google Scholar]
  49. Scheff SW, Price DA, Hicks RR, Baldwin SA, Robinson S, Brackney C. Synaptogenesis in the hippocampal CA1 field following traumatic brain injury. J Neurotrauma. 2005;22(7):719–732. doi: 10.1089/neu.2005.22.719. [DOI] [PubMed] [Google Scholar]
  50. Shen LH, Li Y, Chen J, Zacharek A, Gao Q, Kapke A, Lu M, Raginski K, Vanguri P, Smith A, Chopp M. Therapeutic benefit of bone marrow stromal cells administered 1 month after stroke. J Cereb Blood Flow Metab. 2007;27(1):6–13. doi: 10.1038/sj.jcbfm.9600311. [DOI] [PubMed] [Google Scholar]
  51. Smart N, Risebro CA, Melville AA, Moses K, Schwartz RJ, Chien KR, Riley PR. Thymosin beta4 induces adult epicardial progenitor mobilization and neovascularization. Nature. 2007;445(7124):177–182. doi: 10.1038/nature05383. [DOI] [PubMed] [Google Scholar]
  52. Smith JM, Lunga P, Story D, Harris N, Le Belle J, James MF, Pickard JD, Fawcett JW. Inosine promotes recovery of skilled motor function in a model of focal brain injury. Brain. 2007;130(Pt 4):915–925. doi: 10.1093/brain/awl393. [DOI] [PubMed] [Google Scholar]
  53. Stoica B, Byrnes K, Faden AI. Multifunctional drug treatment in neurotrauma. Neurotherapeutics. 2009;6(1):14–27. doi: 10.1016/j.nurt.2008.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Sun D, Mcginn MJ, Zhou Z, Harvey HB, Bullock MR, Colello RJ. Anatomical integration of newly generated dentate granule neurons following traumatic brain injury in adult rats and its association to cognitive recovery. Exp Neurol. 2007;204(1):264–272. doi: 10.1016/j.expneurol.2006.11.005. [DOI] [PubMed] [Google Scholar]
  55. Sun W, Kim H. Neurotrophic roles of the beta-thymosins in the development and regeneration of the nervous system. Ann N Y Acad Sci. 2007;1112:210–218. doi: 10.1196/annals.1415.013. [DOI] [PubMed] [Google Scholar]
  56. Walker PA, Harting MT, Jimenez F, Shah SK, Pati S, Dash PK, Cox CS., Jr Direct intrathecal implantation of mesenchymal stromal cells leads to enhanced neuroprotection via an NFkappaB-mediated increase in interleukin-6 production. Stem Cells Dev. 2010;19(6):867–876. doi: 10.1089/scd.2009.0188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Wang L, Chopp M, Gregg SR, Zhang RL, Teng H, Jiang A, Feng Y, Zhang ZG. Neural progenitor cells treated with EPO induce angiogenesis through the production of VEGF. J Cereb Blood Flow Metab. 2008;28(7):1361–1368. doi: 10.1038/jcbfm.2008.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Wang L, Zhang Z, Wang Y, Zhang R, Chopp M. Treatment of stroke with erythropoietin enhances neurogenesis and angiogenesis and improves neurological function in rats. Stroke. 2004;35(7):1732–1737. doi: 10.1161/01.STR.0000132196.49028.a4. [DOI] [PubMed] [Google Scholar]
  59. Wible EF, Laskowitz DT. Statins in traumatic brain injury. Neurotherapeutics. 2010;7(1):62–73. doi: 10.1016/j.nurt.2009.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Wu H, Lu D, Jiang H, Xiong Y, Qu C, Li B, Mahmood A, Zhou D, Chopp M. Increase in phosphorylation of Akt and its downstream signaling targets and suppression of apoptosis by simvastatin after traumatic brain injury. J Neurosurg. 2008a;109(4):691–698. doi: 10.3171/JNS/2008/109/10/0691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Wu H, Lu D, Jiang H, Xiong Y, Qu C, Li B, Mahmood A, Zhou D, Chopp M. Simvastatin-mediated upregulation of VEGF and BDNF, activation of the PI3K/Akt pathway, and increase of neurogenesis are associated with therapeutic improvement after traumatic brain injury. J Neurotrauma. 2008b;25(2):130–139. doi: 10.1089/neu.2007.0369. [DOI] [PubMed] [Google Scholar]
  62. Xiong Y, Lu D, Qu C, Goussev A, Schallert T, Mahmood A, Chopp M. Effects of erythropoietin on reducing brain damage and improving functional outcome after traumatic brain injury in mice. J Neurosurg. 2008;109(3):510–521. doi: 10.3171/JNS/2008/109/9/0510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Xiong Y, Mahmood A, Meng Y, Zhang Y, Qu C, Schallert T, Chopp M. Delayed administration of erythropoietin reducing hippocampal cell loss, enhancing angiogenesis and neurogenesis, and improving functional outcome following traumatic brain injury in rats: comparison of treatment with single and triple dose. J Neurosurg. 2010a;113(3):598–608. doi: 10.3171/2009.9.JNS09844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Xiong Y, Mahmood A, Meng Y, Zhang Y, Zhang ZG, Morris DC, Chopp M. Treatment of traumatic brain injury with thymosin beta(4) in rats. J Neurosurg. 2010b doi: 10.3171/2010.4.JNS10118. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Xiong Y, Qu C, Mahmood A, Liu Z, Ning R, Li Y, Kaplan DL, Schallert T, Chopp M. Delayed transplantation of human marrow stromal cell-seeded scaffolds increases transcallosal neural fiber length, angiogenesis, and hippocampal neuronal survival and improves functional outcome after traumatic brain injury in rats. Brain Res. 2009;1263:183–191. doi: 10.1016/j.brainres.2009.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Yamanaka S. Strategies and new developments in the generation of patient-specific pluripotent stem cells. Cell Stem Cell. 2007;1(1):39–49. doi: 10.1016/j.stem.2007.05.012. [DOI] [PubMed] [Google Scholar]
  67. Yang XT, Bi YY, Feng DF. From the vascular microenvironment to neurogenesis. Brain Res Bull. 2010 doi: 10.1016/j.brainresbull.2010.09.008. in press. [DOI] [PubMed] [Google Scholar]
  68. Yoshimura S, Teramoto T, Whalen MJ, Irizarry MC, Takagi Y, Qiu J, Harada J, Waeber C, Breakefield XO, Moskowitz MA. FGF-2 regulates neurogenesis and degeneration in the dentate gyrus after traumatic brain injury in mice. J Clin Invest. 2003;112(8):1202–1210. doi: 10.1172/JCI16618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Zhang J, Zhang ZG, Morris D, Li Y, Roberts C, Elias SB, Chopp M. Neurological functional recovery after thymosin beta4 treatment in mice with experimental auto encephalomyelitis. Neuroscience. 2009a;164(4):1887–1893. doi: 10.1016/j.neuroscience.2009.09.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Zhang Y, Xiong Y, Mahmood A, Meng Y, Liu Z, Qu C, Chopp M. Sprouting of corticospinal tract axons from the contralateral hemisphere into the denervated side of the spinal cord is associated with functional recovery in adult rat after traumatic brain injury and erythropoietin treatment. Brain Res. 2010;1353:249–257. doi: 10.1016/j.brainres.2010.07.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Zhang Y, Xiong Y, Mahmood A, Meng Y, Qu C, Schallert T, Chopp M. Therapeutic effects of erythropoietin on histological and functional outcomes following traumatic brain injury in rats are independent of hematocrit. Brain Res. 2009b;1294:153–164. doi: 10.1016/j.brainres.2009.07.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Zhang ZG, Chopp M. Neurorestorative therapies for stroke: underlying mechanisms and translation to the clinic. Lancet Neurol. 2009;8(5):491–500. doi: 10.1016/S1474-4422(09)70061-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Zhang ZX, Guan LX, Zhang K, Zhang Q, Dai LJ. A combined procedure to deliver autologous mesenchymal stromal cells to patients with traumatic brain injury. Cytotherapy. 2008;10(2):134–139. doi: 10.1080/14653240701883061. [DOI] [PubMed] [Google Scholar]
  74. Zhao C, Deng W, Gage FH. Mechanisms and functional implications of adult neurogenesis. Cell. 2008;132(4):645–660. doi: 10.1016/j.cell.2008.01.033. [DOI] [PubMed] [Google Scholar]

RESOURCES