Current advances in neurotrauma research: diagnosis, neuroprotection, and neurorepair

2014-01-22 16:46JinhuiChen,RiyiShi
中国神经再生研究(英文版) 2014年11期
关键词:网络层应用服务应用层

Current advances in neurotrauma research: diagnosis, neuroprotection, and neurorepair

Traumatic brain injury (TBI) and spinal cord injury (SCI) causes signi fi cant cell death (Raghupathi et al., 1995; DeKosky et al., 1998; Hall et al., 2005; Farkas and Povlishock, 2007) and tissue lesion in the neocortex (Lighthall et al., 1989; Lyeth et al., 1990), leaving many patients with substantial motor disability and cognitive impairment (Hamm et al., 1992; Scheff et al., 1997). Unfortunately, at present, there are no clinically demonstrated FDA approved drug therapies for treatment of TBI and SCI patients that reduce the neurological injuries. Thus, TBI and SCI are serious health problems. The development of therapeutic approaches to prevent neuronal death and enhance neuroregeneration for promoting post-traumatic functional recovery would be of enormous clinical, social, and economic bene fi ts. The reviews in this speci fi c issue focus largely on the current progress on diagnosis, neuroprotection, and potential neurorepair with stem cells.

Introduction

TBI, a form of acquired brain injury, occurs when a sudden trauma causes damage to the brain. TBI can result when the head suddenly and violently hits an object, or when an object pierces the skull and enters brain tissue. It is estimated that approximately 2.4 million patients were hospitalized with TBI in the United states alone in 2009 (Coronado et al., 2012). TBI is a major cause of death and disability in the United States, contributing to about 30% of all injury deaths and the growing 1.8% of the population that live with longterm physical disabilities (Zaloshnja et al., 2008). Effects of TBI can also lead to cognitive impairment, including memory problems and decreased concentration skills, and psychological symptoms, including irritability, depression, and anxiety. SCI is also one of the major causes of irreversible nerve injury, resulting in both motor and sensory dysfunctions. An estimated 12,000 new cases of spinal cord injury occur every year in the United States.

Over the past 15-20 years, we have gained a great deal of knowledge about the healthy brain and its response to trauma (Buki and Povlishock, 2006; Hall et al., 2008; Greer et al., 2013; Johnson et al., 2013). Based on the results from animal models, controlling brain swelling and intracranial pressure (ICP) have been widely used and have signi fi cantly reduced death following TBI (Lundberg et al., 1965). Although during 2001-2010 rates of TBI-related emergency department (ED) visits increased by 70%, death rates decreased by 7% (Coronado et al., 2012).

However, there are still so many questions unanswered, and still so many challenges to diagnose, treat, and repair the damaged brain. To address these challenges, it is very important to advance the knowledge on mechanisms of injury and recovery, and to develop better diagnostic tools and more effective treatments. Thus in this special issue, four laboratories come together to summarize the current progress on neuroimaging, neuroprotection, and potential neurorepair with stem cells following TBI.

Neuroimaging

Different imaging strategies are widely used in the clinic to assess TBI (McAllister et al., 2001; Belanger et al., 2007; Le and Gean, 2009; Kirov et al., 2013). In general, the structural imaging techniques play a role in acute diagnosis and management, while the functional imaging techniques show promise for clari fi cation of pathophysiology, symptom genesis, and mechanisms of recovery (McAllister et al., 2001). Dr. Kuo and Dr. Iraji summarize the most recent evidence of brain plasticity after TBI in human patients from the perspective of advanced magnetic resonance imaging.

Evidence also demonstrates that, even if patients have damaged certain functional structures or networks, e.g., motor control and somatosensory networks, many of them still could pick up these functionalities during their recovery, indicating the existence of an internal neuroplasticity. Dr. Kuo and his colleagues review the most recent imaging evidence of brain plasticity in TBI patients, from synaptic, microstructural levels, to functional network levels of the brain, particularly focusing on advanced MRI.

Neuroprotection

TBI not only results in immediate brain tissue disruption (primary injury), but also causes secondary damage among the surviving cells via complex mechanisms triggered by the primary event occurring in the hours, days, and weeks after initial physical impact. Secondary injury includes ischemia/ reperfusion injury, in fl ammation, oxidative stress, and glutamate excitotoxicity, all of which contribute to the eventual tissue degeneration and functional loss.

A prevalent hypothesis is that TBI increases extracellular levels of the excitatory neurotransmitters such as glutamate (Choi, 1985, 1987, 1988; Braughler and Hall, 1989; Miller et al., 1990; Choi, 1992; Juurlink and Paterson, 1998; Hall and Springer, 2004; Yi and Hazell, 2006). Glutamate, in turn, causes excessive stimulation of N-methyl-D-aspartic acid receptors (NMDA), thus mediating calcium over in fl ux and triggering rapid excitotoxic necrosis that results in traumatic damage to the central nervous system (CNS). For patients who have experienced TBI, no specific pharmacological therapy is available that would improve their outcomes. Therefore, recent research on TBI has been focused on developing a therapeutic approach to inhibit glutamate-mediated excitotoxicity with pharmacological glutamate antagonists or calcium blocking agents. However, glutamate is the major excitatory transmitter in the mammalian CNS. Its stimulation of NMDA receptors plays an essential role in excitatory synaptic transmission. Completely blocking NMDA receptors will cause signi fi cant side effects. For this reason, clinical trials have had limited success.

Another hallmark of secondary injury is oxidative stress (Hall et al., 1999; Bains and Hall, 2012), which plays an important role in mediating functional loss after both TBI and SCI. Although there is strong evidence that oxidative stress plays a critical role in the pathogenesis after SCI, clinical trials of free radical scavenging have not produced any effective treatments to promote functional recovery after traumatic SCI. Dr. Shi and his colleagues found that Acrolein is the most reactive electrophile produced by lipid peroxidation, suggesting that Acrolein is a novel therapeutic target to reduce oxidative stress (Shi et al., 2011a, 2011b; Park et al., 2014). Dr Shi and his colleagues summarize the recent devel-opments in the understanding of the mechanisms of Acrolein in motor and sensory dysfunction in animal models of SCI.

Neurorepair

Recent research has identified neural stem/progenitor cells (NSCs) in the adult mammalian hippocampus that can support neurogenesis throughout life, as demonstrated in rodents and primates, including humans (Kuhn et al., 1996; Eriksson et al., 1998b; Eriksson et al., 1998a; Kornack and Rakic, 1999; Cameron and McKay, 2001; Leuner et al., 2007). Currently the consensus among researchers in the fi eld is that throughout adulthood, NSCs in the subgranular zone (SGZ) of the hippocampal dentate gyrus (HDG) continuously generate new neurons (Kempermann and Gage, 2000; Ming and Song, 2005) and develop into mature granular neurons (Ming and Song, 2005; Shapiro and Ribak, 2005; Zhao et al., 2006). The pool of NSCs is a potential resource for repairing the damaged hippocampus following TBI.

Current studies further show that TBI promotes NSC proliferation in the adult hippocampus (Dash et al., 2001; Kernie et al., 2001; Braun et al., 2002; Chirumamilla et al., 2002; Rice et al., 2003; Yoshimura et al., 2003; Ramaswamy et al., 2005; Sun et al., 2005; Rola et al., 2006; Sun et al., 2007). This finding suggests that innate repair and/or plasticity mechanisms exist in the adult brain. There are distinct classes of NSCs in the adult HDG, including quiescent neural progenitors (QNPs), which carry stem cell properties, and their progeny, amplifying neural progenitors (ANPs) (Seri and Garcia-Verdugo, 2001; Seaberg and van der Kooy, 2002; Filippov et al., 2003; Mignone et al., 2004; Bull and Bartlett, 2005; Encinas et al., 2006; Encinas and Enikolopov, 2008; Encinas et al., 2008). Dr. Chen and his colleagues found that moderate TBI promotes proliferation of QNPs in the adult hippocampus (Gao et al., 2009).

Although TBI promotes NSC proliferation, the effect of TBI on neurogenesis is still controversial. There are con fl icting reports about neurogenesis in the HDG. According to some studies neurogenesis decreases after TBI (Braun et al., 2002; Rola et al., 2006), whereas others have reported that it remains unchanged (Chirumamilla et al., 2002; Rice et al., 2003), or that it increases (Sun et al., 2005; Sun et al., 2007). Here, Dr. Sun summarizes the potential of endogenous neurogenesis for brain repair and regeneration in the hippocampus following traumatic brain injury.

TBI causes signi fi cant cell death (Raghupathi et al., 1995; DeKosky et al., 1998; Hall et al., 2005; Farkas and Povlishock, 2007) and tissue lesion in the neocortex (Lighthall et al., 1989; Lyeth et al., 1990). However, it is generally agreed that no endogenous NSCs exist or neurogenesis proceeds in the adult neocortex of the mammalian brain, i.e., the neocortex is a non-neurogenic region (Rakic, 2006). Thus, Dr. Chen and his colleagues brie fl y review the current progress of stem cells, which may potentially be used to generate new neurons in the cortex for brain repair following TBI.

Summary and future research

Little can be done to reverse the initial brain damage and spinal cord injury caused by trauma. Thus, it is important to study the pathological basis of neurological disorders, understand neurodegeneration and plasticity of the CNS, and develop novel neuroprotection and repair strategies to improve anatomical reorganization and functional recovery following TBI and SCI.

Jinhui Chen1, Riyi Shi2,3

1 Department of Neurological Surgery, Stark Neuroscience

Research Institute, Department of Anatomy, Indiana University School of Medicine, Indianapolis, IN, USA

2 Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA

3 Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA

Bains M, Hall ED (2012) Antioxidant therapies in traumatic brain and spinal cord injury. Biochim Biophys Acta 1822:675-684.

Belanger HG, Vanderploeg RD, Curtiss G, Warden DL (2007) Recent neuroimaging techniques in mild traumatic brain injury. J Neuropsychiatry Clin Neurosci 19:5-20.

Braughler JM, Hall ED (1989) Central nervous system trauma and stroke. I. Biochemical considerations for oxygen radical formation and lipid peroxidation. Free Radic Biol Med 6:289-301.

Braun H, Schafer K, Hollt V (2002) BetaIII tubulin-expressing neurons reveal enhanced neurogenesis in hippocampal and cortical structures after a contusion trauma in rats. J Neurotrauma 19:975-983.

Buki A, Povlishock JT (2006) All roads lead to disconnection?--Traumatic axonal injury revisited. Acta Neurochir (Wien) 148:181-193; discussion 193-184.

Bull ND, Bartlett PF (2005) The adult mouse hippocampal progenitor is neurogenic but not a stem cell. J Neurosci 25:10815-10821.

Cameron HA, McKay RD (2001) Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus. J Comp Neurol 435: 406-417.

Chirumamilla S, Sun D, Bullock MR, Colello RJ (2002) Traumatic brain injury induced cell proliferation in the adult mammalian central nervous system. J Neurotrauma 19:693-703.

Choi DW (1985) Glutamate neurotoxicity in cortical cell culture is calcium dependent. Neurosci Lett 58:293-297.

Choi DW (1987) Ionic dependence of glutamate neurotoxicity. J Neurosci 7:369-379.

Choi DW (1988) Glutamate neurotoxicity and diseases of the nervous system. [Review]. Neuron 1:623-634.

Choi DW (1992) Excitotoxic cell death. J Neurobiol 23:1261-1276.

Coronado VG, McGuire LC, Sarmiento K, Bell J, Lionbarger MR, Jones CD, Geller AI, Khoury N, Xu L (2012) Trends in traumatic brain injury in the U.S. and the public health response: 1995-2009. J Safety Res 43:299-307.

Dash PK, Mach SA, Moore AN (2001) Enhanced neurogenesis in the rodent hippocampus following traumatic brain injury. J Neurosci Res 63:313-319.

DeKosky ST, Kochanek PM, Clark RS, Ciallella JR, Dixon CE (1998) Secondary injury after head trauma: subacute and long-term mechanisms. Semin Clin Neuropsychiatry 3:176-185.

Encinas JM, Enikolopov G (2008) Identifying and quantitating neural stem and progenitor cells in the adult brain. Methods Cell Biol 85: 243-272.

Encinas JM, Vaahtokari A, Enikolopov G (2006) Fluoxetine targets early progenitor cells in the adult brain. Proc Natl Acad Sci U S A 103: 8233-8238.

Encinas JM, Vazquez ME, Switzer RC, Chamberland DW, Nick H, Levine HG, Scarpa PJ, Enikolopov G, Steindler DA (2008) Quiescent adult neural stem cells are exceptionally sensitive to cosmic radiation. Exp Neurol 210:274-279.

Eriksson PS, Per fi lieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, Gage FH (1998a) Neurogenesis in the adult human hippocampus. Nat Med 4:1313-1317.

Eriksson PS, Per fi lieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, Gage FH (1998b) Neurogenesis in the adult human hippocampus. Nat Med 4:1313-1317.

Farkas O, Povlishock JT (2007) Cellular and subcellular change evoked by diffuse traumatic brain injury: a complex web of change extending far beyond focal damage. Prog Brain Res 161:43-59.

Filippov V, Kronenberg G, Pivneva T, Reuter K, Steiner B, Wang LP, Yamaguchi M, Kettenmann H, Kempermann G (2003) Subpopulation of nestin-expressing progenitor cells in the adult murine hippocampus shows electrophysiological and morphological characteristics of astrocytes. Mol Cell Neurosci 23:373-382.

Gao X, Enikolopov G, Chen J (2009) Moderate traumatic brain injury promotes proliferation of quiescent neural progenitors in the adult hippocampus. Exp Neurol 219:516-523.

Greer JE, Hanell A, McGinn MJ, Povlishock JT (2013) Mild traumatic brain injury in the mouse induces axotomy primarily within the axon initial segment. Acta Neuropathol 126:59-74.

Hall ED, Springer JE (2004) Neuroprotection and acute spinal cord injury: a reappraisal. NeuroRx 1:80-100.

Hall ED, Kupina NC, Althaus JS (1999) Peroxynitrite scavengers for the acute treatment of traumatic brain injury. Ann N Y Acad Sci 890: 462-468.

Hall ED, Bryant YD, Cho W, Sullivan PG (2008) Evolution of post-traumatic neurodegeneration after controlled cortical impact traumatic brain injury in mice and rats as assessed by the de Olmos silver and fl uorojade staining methods. J Neurotrauma 25:235-247.

Hall ED, Sullivan PG, Gibson TR, Pavel KM, Thompson BM, Scheff SW (2005) Spatial and temporal characteristics of neurodegeneration after controlled cortical impact in mice: more than a focal brain injury. J Neurotrauma 22:252-265.

Hamm RJ, Dixon CE, Gbadebo DM, Singha AK, Jenkins LW, Lyeth BG, Hayes RL (1992) Cognitive de fi cits following traumatic brain injury produced by controlled cortical impact. J Neurotrauma 9:11-20.

物联网的技术架构分为3层:感知层、网络层和应用层,如图1所示。其中,感知层实现物联网全面智能化感知,网络层将实现接入信息管理和由计算机网络及通信网络构成的承载网络,应用层实现应用支撑服务和用户应用服务。

Johnson VE, Stewart W, Smith DH (2013) Axonal pathology in traumatic brain injury. Exp Neurol 246:35-43.

Juurlink BH, Paterson PG (1998) Review of oxidative stress in brain and spinal cord injury: suggestions for pharmacological and nutritional management strategies. J Spinal Cord Med 21:309-334.

Kempermann G, Gage FH (2000) Neurogenesis in the adult hippocampus. Novartis Found Symp 231:220-235; discussion 235-241, 302-226.

Kernie SG, Erwin TM, Parada LF (2001) Brain remodeling due to neuronal and astrocytic proliferation after controlled cortical injury in mice. J Neurosci Res 66:317-326.

Kirov, II, Tal A, Babb JS, Reaume J, Bushnik T, Ashman TA, Flanagan S, Grossman RI, Gonen O (2013) Proton MR spectroscopy correlates diffuse axonal abnormalities with post-concussive symptoms in mild traumatic brain injury. J Neurotrauma 30:1200-1204.

Kornack DR, Rakic P (1999) Continuation of neurogenesis in the hippocampus of the adult macaque monkey. Proc Natl Acad Sci U S A 96:5768-5773.

Kuhn HG, Dickinson-Anson H, Gage FH (1996) Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci 16:2027-2033.

Le TH, Gean AD (2009) Neuroimaging of traumatic brain injury. Mt Sinai J Med 76:145-162.

Lighthall JW, Dixon CE, Anderson TE (1989) Experimental models of brain injury. J Neurotrauma 6:83-97.

Lundberg N, Troupp H, Lorin H (1965) Continuous recording of the ventricular-fluid pressure in patients with severe acute traumatic brain injury. A preliminary report. J Neurosurg 22:581-590.

Lyeth BG, Jenkins LW, Hamm RJ, Dixon CE, Phillips LL, Clifton GL, Young HF, Hayes RL (1990) Prolonged memory impairment in the absence of hippocampal cell death following traumatic brain injury in the rat. Brain Res 526:249-258.

McAllister TW, Sparling MB, Flashman LA, Saykin AJ (2001) Neuroimaging fi ndings in mild traumatic brain injury. J Clin Exp Neuropsychol 23:775-791.

Mignone JL, Kukekov V, Chiang AS, Steindler D, Enikolopov G (2004) Neural stem and progenitor cells in nestin-GFP transgenic mice. J Comp Neurol 469:311-324.

Miller LP, Lyeth BG, Jenkins LW, Oleniak L, Panchision D, Hamm RJ, Phillips LL, Dixon CE, Clifton GL, Hayes RL (1990) Excitatory amino acid receptor subtype binding following traumatic brain injury. Brain Res 526:103-107.

Ming GL, Song H (2005) Adult neurogenesis in the mammalian central nervous system. Annu Rev Neurosci 28:223-250.

Park J, Zheng L, Marquis A, Walls M, Duerstock B, Pond A, Vega-Alvarez S, Wang H, Ouyang Z, Shi R (2014) Neuroprotective role of hydralazine in rat spinal cord injury-attenuation of acrolein-mediated damage. J Neurochem 129:339-349.

Raghupathi R, McIntosh TK, Smith DH (1995) Cellular responses to experimental brain injury. Brain Pathol 5:437-442.

Rakic P (2006) Neuroscience. No more cortical neurons for you. Science 313:928-929.

Ramaswamy S, Goings GE, Soderstrom KE, Szele FG, Kozlowski DA (2005) Cellular proliferation and migration following a controlled cortical impact in the mouse. Brain Res 1053:38-53.

Rice AC, Khaldi A, Harvey HB, Salman NJ, White F, Fillmore H, Bullock MR (2003) Proliferation and neuronal differentiation of mitotically active cells following traumatic brain injury. Exp Neurol 183:406-417. Rola R, Mizumatsu S, Otsuka S, Morhardt DR, Noble-Haeusslein LJ, Fishman K, Potts MB, Fike JR (2006) Alterations in hippocampal neurogenesis following traumatic brain injury in mice. Exp Neurol 202:189-199.

Scheff SW, Baldwin SA, Brown RW, Kraemer PJ (1997) Morris water maze deficits in rats following traumatic brain injury: lateral controlled cortical impact. J Neurotrauma 14:615-627.

Seaberg RM, van der Kooy D (2002) Adult rodent neurogenic regions: the ventricular subependyma contains neural stem cells, but the dentate gyrus contains restricted progenitors. J Neurosci 22:1784-1793.

Seri B, Garcia-Verdugo JM (2001) Astrocytes give rise to new neurons in the adult mammalian hippocampus. J Neurosci 21:7153-7160.

Shapiro LA, Ribak CE (2005) Integration of newly born dentate granule cells into adult brains: hypotheses based on normal and epileptic rodents. Brain Res Brain Res Rev 48:43-56.

Shi R, Rickett T, Sun W (2011a) Acrolein-mediated injury in nervous system trauma and diseases. Mol Nutr Food Res 55:1320-1331.

Shi Y, Sun W, McBride JJ, Cheng JX, Shi R (2011b) Acrolein induces myelin damage in mammalian spinal cord. J Neurochem 117:554-564.

Sun D, McGinn MJ, Zhou Z, Harvey HB, Bullock MR, Colello RJ (2007) Anatomical integration of newly generated dentate granule neurons following traumatic brain injury in adult rats and its association to cognitive recovery. Exp Neurol 204:264-272.

Sun D, Colello RJ, Daugherty WP, Kwon TH, McGinn MJ, Harvey HB, Bullock MR (2005) Cell proliferation and neuronal differentiation in the dentate gyrus in juvenile and adult rats following traumatic brain injury. J Neurotrauma 22:95-105.

Yi JH, Hazell AS (2006) Excitotoxic mechanisms and the role of astrocytic glutamate transporters in traumatic brain injury. Neurochem Int 48:394-403.

Yoshimura S, Teramoto T, Whalen MJ, Irizarry MC, Takagi Y, Qiu J, Harada J, Waeber C, Breake fi eld XO, Moskowitz MA (2003) FGF-2 regulates neurogenesis and degeneration in the dentate gyrus after traumatic brain injury in mice. J Clin Invest 112:1202-1210.

Zaloshnja E, Miller T, Langlois JA, Selassie AW (2008) Prevalence of long-term disability from traumatic brain injury in the civilian population of the United States, 2005. J Head Trauma Rehabil 23:394-400.

Zhao C, Teng EM, Summers RG, Jr., Ming GL, Gage FH (2006) Distinct morphological stages of dentate granule neuron maturation in the adult mouse hippocampus. J Neurosci 26:3-11.

Jinhui Chen, M.D., Ph.D., Department of Neurological Surgery, Stark Neuroscience Research Institute,

10.4103/1673-5374.135306 http://www.nrronline.org/

Department of Anatomy, Indiana University School of Medicine, Indianapolis, IN 46202, USA, chen204@iupui.edu. Riyi Shi, M.D., Ph.D., Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA, riyi@purdue.edu.

Accepted: 2014-06-05

Chen J, Shi R. Current advances in neurotrauma research: diagnosis, neuroprotection, and neurorepair. Neural Regen Res. 2014;9(11):1093-1095.

猜你喜欢
网络层应用服务应用层
加快推进航天强国建设 提升卫星应用服务能力——两会代表委员为航天发展建言献策
全球卫星互联网应用服务及我国的发展策略
论物联网安全威胁及其应对策略
传输层和应用层的隧道技术
基于分级保护的OA系统应用层访问控制研究
物联网技术在信息机房制冷系统中的应用
全国征集卫星应用服务解决方案
微软推出整合版Azure应用服务
浅析TDCS/CTC系统中心网络防火墙的链接