簡易檢索 / 詳目顯示

研究生: 趙伯寬
Po-Kuan Chao
論文名稱: 以大鼠周邊及中樞神經損傷模式探討調節細胞激素對於神經系統的保護機制
Elucidate the Neural Protective Mechanism of Regulation of Cytokines on the Peripheral Nerve Injury and Central Neuronal Damage Models in Rat
指導教授: 呂國棟
Lu, Kwok-Tung
羅榮昇
Ro, Long-Sun
楊奕玲
Yang, Yi-Ling
學位類別: 博士
Doctor
系所名稱: 生命科學系
Department of Life Science
論文出版年: 2012
畢業學年度: 100
語文別: 英文
論文頁數: 97
中文關鍵詞: 顆粒細胞刺激增生因子腫瘤壞死因子小膠細胞神經損傷神經病變痛鴉片類物質細胞激素
英文關鍵詞: granulocyte colony-stimulating factor, tumor necrosis factor, microglia, nerve injury, neuropathic pain, opioid, cytokine
論文種類: 學術論文
相關次數: 點閱:338下載:2
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報
  • 近年來的研究顯示,調節細胞激素有助於減少神經損傷後的神經發炎並且改善神經系統功能。在本研究當中,我們分別使用不同的大鼠模式來研究如何透過特定調節物質來改變周邊神經損傷及中樞神經傷害後細胞激素的表現。
    首先,為了釐清組織病理評估結果與行為結果之間的關聯性,我們針對假手術對照組及長期壓迫神經損傷手術組大鼠的同側及對側脊髓神經背角及第五段腰椎的背根神經結,分別測量其c-Fos蛋白質表現以及小膠細胞活化作用。我們發現c-Fos蛋白質表現在長期壓迫神經損傷手術組大鼠的同側背根神經結並不會增加。此外,手術後72小時長期壓迫神經損傷引發c-Fos蛋白質表現增加只會出現在同側脊髓神經背角中。然而相對的,小膠細胞活化現象在脊髓神經背角兩側都非常顯著,同時也與機械性刺激引發痛覺過敏的行為結果一致。所以我們證實不僅有神經細胞、小膠細胞也在神經病變痛當中扮演角色,在中樞神經系統中,小膠細胞透過與神經細胞之間的交互作用對疼痛行為產生影響。
    先前的研究中發現在神經系統中顆粒細胞刺激增生因子是一種重要的調節物質。所以本研究展示了與對照組相較的結果,在手術後的1到25天之間,投予顆粒細胞刺激增生因子有助於減輕長期壓迫神經損傷引發的熱痛覺過敏及機械性刺激過敏的現象。長期壓迫神經損傷手術手術後投予顆粒細胞刺激增生因子也減少動物熱痛覺過敏。推測若神經損傷後48小時內單次投與顆粒細胞刺激增生因子仍對於神經病變痛的治療具有效果。顆粒細胞刺激增生因子也驅動含鴉片類物質的聚多核細胞聚集進入受損的神經之中。長期壓迫神經損傷不僅導致背根神經結中的第六型細胞激素的mRNA及腫瘤壞死因子α亞型蛋白表現量增加,也使得脊髓中第一型細胞激素的蛋白表現量上升。這些細胞激素的產生都會被顆粒細胞刺激增生因子所抑制。我們更進一步發現長期壓迫神經損傷後,μ型鴉片受體出現在損傷的神經當中而且鴉片受體拮抗劑─naloxone methiodide減低了顆粒細胞刺激增生因子的抗痛效果,於是我們推論顆粒細胞刺激增生因子減輕熱痛覺過敏是透過鴉片類物質及其受體之間交互作用的結果。更進一步來看,投與顆粒細胞刺激增生因子可以抑制脊髓神經背角處因長期壓迫神經損傷所引發小膠細胞大量活化。這些結果推測在神經損傷初期單次全身性投予顆粒細胞刺激增生因子減輕神經病變痛的效果是透過活化聚多核細胞驅動的內生性鴉片類物質分泌,進而活化損傷神經當中的鴉片類物質受體,同時減少促發炎細胞激素,並減輕脊髓神經背角當中的小膠細胞活化現象。
    而在大鼠神經損傷模式中,indomethacin曾被用來進行創傷的治療。基於西方墨漬法的分析結果, 我們發現Nogo-A蛋白表現在頭部創傷後八小時顯著增加。此外,頭部創傷引發受測動物的第一型細胞激素含量顯著提升,一般而言所有的頭部創傷相關的分子及細胞的後續反應都會被抗發炎藥物indomethacin所影響而顯著減少。更重要的是,頭部創傷刺激作用相關的Nogo-A及第一型細胞激素的含量明顯被具有專一性的反義寡核苷酸所抑制。我們的發現,出現Nogo-A表現受到抑制的初期反應是由indomethacin所賦予的,也減少第一型細胞激素的表現程度並且減輕頭部誘發的神經損傷。
    總結以上,由於全身性投予顆粒細胞刺激增生因子或是indomethacin分別對於大鼠的周邊以及中樞神經損傷具有正面的影響,以顆粒細胞刺激增生因子或者是indomethacin來調節中樞神經系統中的促發炎細胞激素,很可能有成為治療中樞神經過敏化現象的嶄新療法。

    Recent studies have shown that modulation of cytokine expression is helpful to reduce nerve inflammation and to improve dysfunction of nerve system. In this study, we use rat models to investigate the mediators for alterations of cytokines expression following peripheral nerve injury and central neural damage.
    First, in order to clarify the correlation between histopathological assessment and behavior outcome, c-Fos protein expression and microglia activation were quantified in both of the ipsi- and contra-lateral spinal dorsal horn (DH) and/or Lumbar 5 (L5) dorsal root ganglia (DRG) in sham-operation and chronic constriction injury (CCI) rats. We found that c-Fos protein expression did not increase in the ipsilateral DRG of CCI rat. In addition, CCI evoked increase of c-Fos protein expression was only in the ipsilateral spinal DH after 72h. In contrast, however, microglia activation was notably induced at bilateral spinal DH and correlated with mechanical allodynia. Thus, neurons are not the only cells playing a role in neuropathic pain, we evidence that microglia are involved as well and there is a cross effect between neuron and microglia in the central nervous system (CNS) which associated to nociceptive behaviors.
    The current studies show that granulocyte colony-stimulating factor (G-CSF) is an important mediator of nerve system. Secondary, this study shows the effectiveness of exogenous treatment for alleviating thermal hyperalgesia and mechanical allodynia in rats with CCI, during post-operative days 1–25, compared to that of vehicle treatment. G-CSF also increases the recruitment of opioid-containing PMN cells into the injured nerve. After CCI, single administration of G-CSF, relieved thermal hyperalgesia, indicated that its effect on neuropathic pain had a therapeutic window of 0–48 h after nerve injury. CCI led to increase in the levels of interleukin-6 (IL-6) mRNA and tumor necrosis factor-alpha (TNF-α) protein in the DRG and interleukin-1beta (IL-1β) protein in the spinal cord. These high levels of IL-6 mRNA, TNF-α and IL-1β were, respectively, suppressed by a single administration of G-CSF after CCI, respectively. Moreover, the μ-opioid receptor was observed in injured nerve and opioid receptor antagonist naloxone methiodide (NLXM) reversed G-CSF-induced antinociception after CCI, suggesting that G-CSF alleviates hyperalgesia via opioid/opioid receptor interactions. Furthermore, G-CSF administered after CCI suppressed the CCI-induced up-regulation of microglial activation in the ipsilateral spinal DH, which is essential for sensing neuropathic pain. These results suggest that an early single systemic injection of G-CSF alleviates neuropathic pain via activation of PMN cell-derived endogenous opioid secretion to activate opioid receptors in the injured nerve, down-regulate IL-6, IL-1β and TNF- pro-inflammatory cytokines, and attenuate microglial activation in the spinal DH.
    In the neuronal damaged models, indomethacin was ever used to deliver a traumatic management to rats. Based on Western blot analyses, the expression of Nogo-A was found to be significantly up-regulated in the hippocampus beginning eight hours after traumatic brain injury (TBI). In addition, TBI caused an apparent elevation in IL-1β levels in the tested animals. All of the TBI-associated molecular and cellular consequences could be effectively reversed by treating the animals with the anti-inflammatory drug indomethacin. More importantly, the TBI-associated stimulation in the levels of both Nogo-A and IL-1β could be effectively inhibited by a specific Nogo-A antisense oligonucleotide. Our findings suggest that the suppression of Nogo-A expression appears to be an early response conferred by indomethacin, which then leads to decreases in the levels of IL-1β and TBI-induced neuron damage.
    In conclusion, because systemic administration of G-CSF or indomethacin had positive effects in both models of peripheral nerve injury and central nervous system injury in rats, respectively, it is highly possible that treating with G-CSF or indomethacin to modulate pro-inflammatory cytokines of CNS is the new method to manage central sensitization.

    Primary Abstract IV Primary Abstract (Chinese) VII Abbreviations X 1. Introduction 1 1.1 Neuropathic pain 1 1.2 Traumatic brain injury 3 1.3 Part I 4 1.4 Part II 8 2. Methods 13 2.1 Experimental animals 13 2.2 Behavioral tests 16 2.3 Blood cell counting and flow cytometry 17 2.4 IL-6 mRNA assay 18 2.5 Western blotting assay 20 2.6 Enzyme-linked immunosorbent assay (ELISA) 21 2.7 Immunohistochemistry 21 2.8 Statistical analysis 23 3. Results 24 3.1 Part I 24 3.2 Part II 28 4. Discussion 36 4.1 Part I 36 4.2 Part II 45 5. Conclusions 55 6. References 58 7. Figures & Legends 71 Figure 1 After unilateral CCI of the sciatic nerve, animals developed mechanical allodynia in both ipsi- and contra-lateral limbs 71 Figure 2 The CCI operation mobilized immune cells into the injured nerves 73 Figure 3 c-Fos protein did not increase in the ipsilateral DRG after peripheral nerve injury 74 Figure 4 c-Fos protein expressed in bilateral spinal DH of CCI rat 75 Figure 5 CCI induces OX-42 positive immunoreactivity in bilateral spinal DH at 72 h after CCI…………………………………………….76 Figure 6 The number of OX-42 immunoreactive cells in the bilateral spinal DH correlated with mechanical paw withdrawal response of CCI rat at 72 h..………………………………………..……….. 78 Figure 7 The number of OX-42 immunoreactive cells in the contralateral spinal DH correlated with ipsilateral side of CCI rat at 72 h…... 79 Figure 8 G-CSF alleviates long-term thermal hyperalgesia and mechanical allodynia in rats with CCI 80 Figure 9 G-CSF demonstrates a therapeutic time window (0–48 h) after nerve injury………………………………………………………82 Figure 10 Effects of G-CSF on mobilized bone marrow cells in the peripheral blood. 83 Figure 11 Effects of G-CSF on mobilized immune cells in the tissue fluid around the injured nerve 84 Figure 12 G-CSF treatment increases the number of migrated β-endorphin-containing PMN cells in injured nerves 86 Figure 13 μ-opioid receptor expressed in the sciatic nerve of both sham-operated and CCI-operated rat 88 Figure 14 NLXM reverses the G-CSF anti-nociceptive effect 89 Figure 15 G-CSF reduces the levels of IL-6 mRNA and TNF-α protein in the DRG after CCI 91 Figure 16 Expression of IL-1β protein is up-regulated in ipsilateral spinal DH of vehicle- treated CCI rat 93 Figure 17 G-CSF reduces immunoreactivity of OX-42 in the spinal cord 72 h after CCI 94 8. Figures and Legends of References 96 Reference figure 1 Modulation of neuropathic pain by immune cells. 96

    Akins PT, McCleskey EW (1993) Characterization of potassium currents in adult rat sensory neurons and modulation by opioids and cyclic AMP. Neuroscience 56:759-769.
    Baron R, Schwarz K, Kleinert A, Schattschneider J, Wasner G (2001) Histamine-induced itch converts into pain in neuropathic hyperalgesia. Neuroreport 12:3475-3478.
    Becker PS, Wagle M, Matous S, Swanson RS, Pihan G, Lowry PA, Stewart FM, Heard SO (1997) Spontaneous splenic rupture following administration of granulocyte colony-stimulating factor (G-CSF): occurrence in an allogeneic donor of peripheral blood stem cells. Biol Blood Marrow Transplant 3:45-49.
    Bennett GJ, Xie YK (1988) A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 33:87-107.
    Berrios I, Castro C, Kuffler DP (2008) Morphine: axon regeneration, neuroprotection, neurotoxicity, tolerance, and neuropathic pain. P R Health Sci J 27:119-128.
    Bester H, Beggs S, Woolf CJ (2000) Changes in tactile stimuli-induced behavior and c-Fos expression in the superficial dorsal horn and in parabrachial nuclei after sciatic nerve crush. The Journal of Comparative Neurology 428:45-61.
    Binder W, Mousa SA, Sitte N, Kaiser M, Stein C, Schafer M (2004) Sympathetic activation triggers endogenous opioid release and analgesia within peripheral inflamed tissue. Eur J Neurosci 20:92-100.
    Brack A, Rittner HL, Machelska H, Beschmann K, Sitte N, Schafer M, Stein C (2004a) Mobilization of opioid-containing polymorphonuclear cells by hematopoietic growth factors and influence on inflammatory pain. Anesthesiology 100:149-157.
    Brack A, Rittner HL, Machelska H, Leder K, Mousa SA, Schafer M, Stein C (2004b) Control of inflammatory pain by chemokine-mediated recruitment of opioid-containing polymorphonuclear cells. Pain 112:229-238.
    Brack A, Rittner HL, Machelska H, Shaqura M, Mousa SA, Labuz D, Zollner C, Schafer M, Stein C (2004c) Endogenous peripheral antinociception in early inflammation is not limited by the number of opioid-containing leukocytes but by opioid receptor expression. Pain 108:67-75.
    Brack A, Rittner HL, Stein C (2004d) Neurogenic painful inflammation. Curr Opin Anaesthesiol 17:461-464.
    Bullitt E, Lee CL, Light AR, Willcockson H (1992) The effect of stimulus duration on noxious-stimulus induced c-fos expression in the rodent spinal cord. Brain Research 580:172-179.
    Busch-Dienstfertig M, Stein C (2010) Opioid receptors and opioid peptide-producing leukocytes in inflammatory pain--basic and therapeutic aspects. Brain Behav Immun 24:683-694.
    Cabot PJ (2001) Immune-derived opioids and peripheral antinociception. Clinical and Experimental Pharmacology & Physiology 28:230-232.
    Cabot PJ, Carter L, Gaiddon C, Zhang Q, Schafer M, Loeffler JP, Stein C (1997) Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. The Journal of Clinical Investigation 100:142-148.
    Cabot PJ, Carter L, Schafer M, Stein C (2001) Methionine-enkephalin-and Dynorphin A-release from immune cells and control of inflammatory pain. Pain 93:207-212.
    Caggiano AO, Kraig RP (1999) Prostaglandin E receptor subtypes in cultured rat microglia and their role in reducing lipopolysaccharide-induced interleukin-1beta production. Journal of Neurochemistry 72:565-575.
    Cernak I (2005) Animal models of head trauma. NeuroRx : the journal of the American Society for Experimental NeuroTherapeutics 2:410-422.
    Cernak I, Noble-Haeusslein LJ (2010) Traumatic brain injury: an overview of pathobiology with emphasis on military populations. Journal of Cerebral Blood Flow and Metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 30:255-266.
    Cernak I, Vink R, Zapple DN, Cruz MI, Ahmed F, Chang T, Fricke ST, Faden AI (2004) 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.
    Chen MS, Huber AB, van der Haar ME, Frank M, Schnell L, Spillmann AA, Christ F, Schwab ME (2000) Nogo-A is a myelin-associated neurite outgrowth inhibitor and an antigen for monoclonal antibody IN-1. Nature 403:434-439.
    Chen T, Liu W, Chao X, Zhang L, Qu Y, Huo J, Fei Z (2011) Salvianolic acid B attenuates brain damage and inflammation after traumatic brain injury in mice. Brain Research Bulletin 84:163-168.
    Colburn RW, DeLeo JA, Rickman AJ, Yeager MP, Kwon P, Hickey WF (1997) Dissociation of microglial activation and neuropathic pain behaviors following peripheral nerve injury in the rat. Journal of Neuroimmunology 79:163-175.
    Costigan M, Scholz J, Woolf CJ (2009) Neuropathic pain: a maladaptive response of the nervous system to damage. Annual Review of Neuroscience 32:1-32.
    Dale CS, Pagano Rde L, Rioli V (2005) Hemopressin: a novel bioactive peptide derived from the alpha1-chain of hemoglobin. Mem Inst Oswaldo Cruz 100 Suppl 1:105-106.
    David S, Fry EJ, Lopez-Vales R (2008) Novel roles for Nogo receptor in inflammation and disease. Trends in Neurosciences 31:221-226.
    Delander GE, Schott E, Brodin E, Fredholm BB (1997) Temporal changes in spinal cord expression of mRNA for substance P, dynorphin and enkephalin in a model of chronic pain. Acta Physiologica Scandinavica 161:509-516.
    DeLeo JA, Tanga FY, Tawfik VL (2004) Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry 10:40-52.
    Detloff MR, Fisher LC, McGaughy V, Longbrake EE, Popovich PG, Basso DM (2008) Remote activation of microglia and pro-inflammatory cytokines predict the onset and severity of below-level neuropathic pain after spinal cord injury in rats. Experimental Neurology 212:337-347.
    Donnelly DJ, Popovich PG (2008) Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Experimental Neurology 209:378-388.
    Dworkin RH, O'Connor AB, Backonja M, Farrar JT, Finnerup NB, Jensen TS, Kalso EA, Loeser JD, Miaskowski C, Nurmikko TJ, Portenoy RK, Rice AS, Stacey BR, Treede RD, Turk DC, Wallace MS (2007) Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain 132:237-251.
    Endres-Becker J, Heppenstall PA, Mousa SA, Labuz D, Oksche A, Schafer M, Stein C, Zollner C (2007) Mu-opioid receptor activation modulates transient receptor potential vanilloid 1 (TRPV1) currents in sensory neurons in a model of inflammatory pain. Molecular Pharmacology 71:12-18.
    Eyles JL, Hickey MJ, Norman MU, Croker BA, Roberts AW, Drake SF, James WG, Metcalf D, Campbell IK, Wicks IP (2008) A key role for G-CSF-induced neutrophil production and trafficking during inflammatory arthritis. Blood 112:5193-5201.
    Fecho K, Manning EL, Maixner W, Schmitt CP (2007) Effects of carrageenan and morphine on acute inflammation and pain in Lewis and Fischer rats. Brain Behav Immun 21:68-78.
    Finley MJ, Happel CM, Kaminsky DE, Rogers TJ (2008) Opioid and nociceptin receptors regulate cytokine and cytokine receptor expression. Cell Immunol 252:146-154.
    Fleming JC, Norenberg MD, Ramsay DA, Dekaban GA, Marcillo AE, Saenz AD, Pasquale-Styles M, Dietrich WD, Weaver LC (2006) The cellular inflammatory response in human spinal cords after injury. Brain : a journal of neurology 129:3249-3269.
    Fournier AE, GrandPre T, Strittmatter SM (2001) Identification of a receptor mediating Nogo-66 inhibition of axonal regeneration. Nature 409:341-346.
    Fry EJ, Ho C, David S (2007) A role for Nogo receptor in macrophage clearance from injured peripheral nerve. Neuron 53:649-662.
    Gao YJ, Ji RR (2010) Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol Ther 126:56-68.
    Gaskin DJ, Richard P (2012) The Economic Costs of Pain in the United States. The Journal of Pain : official journal of the American Pain Society.
    George A, Schmidt C, Weishaupt A, Toyka KV, Sommer C (1999) Serial determination of tumor necrosis factor-alpha content in rat sciatic nerve after chronic constriction injury. Experimental Neurology 160:124-132.
    Gogas KR, Presley RW, Levine JD, Basbaum AI (1991) The antinociceptive action of supraspinal opioids results from an increase in descending inhibitory control: correlation of nociceptive behavior and c-fos expression. Neuroscience 42:617-628.
    Gold MS, Levine JD (1996) DAMGO inhibits prostaglandin E2-induced potentiation of a TTX-resistant Na+ current in rat sensory neurons in vitro. Neuroscience Letters 212:83-86.
    Gosselin RD, Bebber D, Decosterd I (2010) Upregulation of the GABA transporter GAT-1 in the gracile nucleus in the spared nerve injury model of neuropathic pain. Neuroscience Letters 480:132-137.
    Graeber MB (2010) Changing face of microglia. Science 330:783-788.
    GrandPre T, Li S, Strittmatter SM (2002) Nogo-66 receptor antagonist peptide promotes axonal regeneration. Nature 417:547-551.
    GrandPre T, Nakamura F, Vartanian T, Strittmatter SM (2000) Identification of the Nogo inhibitor of axon regeneration as a Reticulon protein. Nature 403:439-444.
    Hagiwara S, Iwasaka H, Hasegawa A, Noguchi T (2008) Pre-Irradiation of blood by gallium aluminum arsenide (830 nm) low-level laser enhances peripheral endogenous opioid analgesia in rats. Anesthesia and Analgesia 107:1058-1063.
    Hanamsagar R, Torres V, Kielian T (2011) Inflammasome activation and IL-1beta/IL-18 processing are influenced by distinct pathways in microglia. Journal of Neurochemistry 119:736-748.
    Hanisch UK, Kettenmann H (2007) Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nature Neuroscience 10:1387-1394.
    Harris JA (1998) Using c-fos as a neural marker of pain. Brain Research Bulletin 45:1-8.
    Hathway GJ, Vega-Avelaira D, Moss A, Ingram R, Fitzgerald M (2009) Brief, low frequency stimulation of rat peripheral C-fibres evokes prolonged microglial-induced central sensitization in adults but not in neonates. Pain 144:110-118.
    Herdegen T, Kovary K, Leah J, Bravo R (1991a) Specific temporal and spatial distribution of JUN, FOS, and KROX-24 proteins in spinal neurons following noxious transsynaptic stimulation. The Journal of Comparative Neurology 313:178-191.
    Herdegen T, Tolle TR, Bravo R, Zieglgansberger W, Zimmermann M (1991b) Sequential expression of JUN B, JUN D and FOS B proteins in rat spinal neurons: cascade of transcriptional operations during nociception. Neuroscience Letters 129:221-224.
    Hoggatt J, Pelus LM (2011) Many mechanisms mediating mobilization: an alliterative review. Curr Opin Hematol 18:231-238.
    Honore P, Buritova J, Besson JM (1995) Carrageenin-evoked c-Fos expression in rat lumbar spinal cord: the effects of indomethacin. European Journal of Pharmacology 272:249-259.
    Hsieh GC, Chandran P, Salyers AK, Pai M, Zhu CZ, Wensink EJ, Witte DG, Miller TR, Mikusa JP, Baker SJ, Wetter JM, Marsh KC, Hancock AA, Cowart MD, Esbenshade TA, Brioni JD, Honore P (2010) H4 receptor antagonism exhibits anti-nociceptive effects in inflammatory and neuropathic pain models in rats. Pharmacology, Biochemistry, and Behavior 95:41-50.
    Huang X, Yang J, Chang JK, Dun NJ (2010) Amylin suppresses acetic acid-induced visceral pain and spinal c-fos expression in the mouse. Neuroscience 165:1429-1438.
    Huber AB, Weinmann O, Brosamle C, Oertle T, Schwab ME (2002) Patterns of Nogo mRNA and protein expression in the developing and adult rat and after CNS lesions. The Journal of Neuroscience : the official journal of the Society for Neuroscience 22:3553-3567.
    Hunt D, Coffin RS, Prinjha RK, Campbell G, Anderson PN (2003) Nogo-A expression in the intact and injured nervous system. Molecular and Cellular Neurosciences 24:1083-1102.
    Inoue K, Tsuda M (2009) Microglia and neuropathic pain. Glia 57:1469-1479.
    Iwasaka H, Kitano T, Miyakawa H, Unoshima M, Shinguu C, Matsumoto S, Noguchi T (2001) Neutrophilia and granulocyte colony-stimulating factor levels after cardiopulmonary bypass. Can J Anaesth 48:81-84.
    Jancalek R, Dubovy P, Svizenska I, Klusakova I (2010) Bilateral changes of TNF-alpha and IL-10 protein in the lumbar and cervical dorsal root ganglia following a unilateral chronic constriction injury of the sciatic nerve. Journal of Neuroinflammation 7:11.
    Jasmin L, Wang H, Tarczy-Hornoch K, Levine JD, Basbaum AI (1994) Differential effects of morphine on noxious stimulus-evoked fos-like immunoreactivity in subpopulations of spinoparabrachial neurons. The Journal of Neuroscience : the official journal of the Society for Neuroscience 14:7252-7260.
    Jergova S, Kolesar D, Cizkova D (2008) Expression of c-Fos in the parabrachial nucleus following peripheral nerve injury in rats. Eur J Pain 12:172-179.
    Ji RR, Baba H, Brenner GJ, Woolf CJ (1999) Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nature Neuroscience 2:1114-1119.
    Jones TB, McDaniel EE, Popovich PG (2005) Inflammatory-mediated injury and repair in the traumatically injured spinal cord. Current Pharmaceutical Design 11:1223-1236.
    Jurgens HA, Johnson RW (2012) Dysregulated neuronal-microglial cross-talk during aging, stress and inflammation. Experimental Neurology 233:40-48.
    Kapitzke D, Vetter I, Cabot PJ (2005) Endogenous opioid analgesia in peripheral tissues and the clinical implications for pain control. Ther Clin Risk Manag 1:279-297.
    Koprivica V, Cho KS, Park JB, Yiu G, Atwal J, Gore B, Kim JA, Lin E, Tessier-Lavigne M, Chen DF, He Z (2005) EGFR activation mediates inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans. Science 310:106-110.
    Kunz T, Marklund N, Hillered L, Oliw EH (2002) Cyclooxygenase-2, prostaglandin synthases, and prostaglandin H2 metabolism in traumatic brain injury in the rat. Journal of Neurotrauma 19:1051-1064.
    Labuz D, Schmidt Y, Schreiter A, Rittner HL, Mousa SA, Machelska H (2009) Immune cell-derived opioids protect against neuropathic pain in mice. The Journal of Clinical Investigation 119:278-286.
    Labuz D, Schreiter A, Schmidt Y, Brack A, Machelska H (2010) T lymphocytes containing beta-endorphin ameliorate mechanical hypersensitivity following nerve injury. Brain Behav Immun 24:1045-1053.
    Lazarini F, Tham TN, Casanova P, Arenzana-Seisdedos F, Dubois-Dalcq M (2003) Role of the alpha-chemokine stromal cell-derived factor (SDF-1) in the developing and mature central nervous system. Glia 42:139-148.
    Leanez S, Hervera A, Pol O (2009) Peripheral antinociceptive effects of micro- and delta-opioid receptor agonists in NOS2 and NOS1 knockout mice during chronic inflammatory pain. European Journal of Pharmacology 602:41-49.
    Lee IO, Seo Y (2008) The effects of intrathecal cyclooxygenase-1, cyclooxygenase-2, or nonselective inhibitors on pain behavior and spinal Fos-like immunoreactivity. Anesthesia and Analgesia 106:972-977, table of contents.
    Lee JK, Kim JE, Sivula M, Strittmatter SM (2004) Nogo receptor antagonism promotes stroke recovery by enhancing axonal plasticity. The Journal of N euroscience : the official journal of the Society for Neuroscience 24:6209-6217.
    Leonardo CC, Hall AA, Collier LA, Gottschall PE, Pennypacker KR (2009) Inhibition of gelatinase activity reduces neural injury in an ex vivo model of hypoxia-ischemia. Neuroscience 160:755-766.
    Leung L, Cahill CM (2010) TNF-alpha and neuropathic pain--a review. Journal of Neuroinflammation 7:27.
    Li X, Lighthall G, Liang DY, Clark JD (2004) Alterations in spinal cord gene expression after hindpaw formalin injection. Journal of Neuroscience Research 78:533-541.
    Liou JT, Lui PW, Liu FC, Lai YS, Day YJ (2011) Exogenous granulocyte colony-stimulating factor exacerbate pain-related behaviors after peripheral nerve injury. J Neuroimmunol 232:83-93.
    Lu KT, Cheng NC, Wu CY, Yang YL (2008) NKCC1-mediated traumatic brain injury-induced brain edema and neuron death via Raf/MEK/MAPK cascade. Critical care medicine 36:917-922.
    Lu KT, Wang YW, Yang JT, Yang YL, Chen HI (2005) Effect of interleukin-1 on traumatic brain injury-induced damage to hippocampal neurons. Journal of Neurotrauma 22:885-895.
    Lynch JR, Wang H, Mace B, Leinenweber S, Warner DS, Bennett ER, Vitek MP, McKenna S, Laskowitz DT (2005) A novel therapeutic derived from apolipoprotein E reduces brain inflammation and improves outcome after closed head injury. Experimental Neurology 192:109-116.
    Machelska H (2011) Dual peripheral actions of immune cells in neuropathic pain. Archivum immunologiae et therapiae experimentalis 59:11-24.
    Machelska H, Heppenstall PA, Stein C (2003) Breaking the pain barrier. Nat Med 9:1353-1354.
    Mackrell PJ, Daly JM, Mestre JR, Stapleton PP, Howe LR, Subbaramaiah K, Dannenberg AJ (2001) Elevated expression of cyclooxygenase-2 contributes to immune dysfunction in a murine model of trauma. Surgery 130:826-833.
    Marchand F, Perretti M, McMahon SB (2005) Role of the immune system in chronic pain. Nature Reviews Neuroscience 6:521-532.
    Markus TM, Tsai SY, Bollnow MR, Farrer RG, O'Brien TE, Kindler-Baumann DR, Rausch M, Rudin M, Wiessner C, Mir AK, Schwab ME, Kartje GL (2005) Recovery and brain reorganization after stroke in adult and aged rats. Annals of Neurology 58:950-953.
    Marmarou A, Foda MA, van den Brink W, Campbell J, Kita H, Demetriadou K (1994) A new model of diffuse brain injury in rats. Part I: Pathophysiology and biomechanics. Journal of Neurosurgery 80:291-300.
    McMahon SB, Cafferty WB, Marchand F (2005) Immune and glial cell factors as pain mediators and modulators. Experimental Neurology 192:444-462.
    Mousa SA, Bopaiah CP, Richter JF, Yamdeu RS, Schafer M (2007) Inhibition of inflammatory pain by CRF at peripheral, spinal and supraspinal sites: involvement of areas coexpressing CRF receptors and opioid peptides. Neuropsychopharmacology 32:2530-2542.
    Mousa SA, Machelska H, Schafer M, Stein C (2002) Immunohistochemical localization of endomorphin-1 and endomorphin-2 in immune cells and spinal cord in a model of inflammatory pain. J Neuroimmunol 126:5-15.
    Mousa SA, Shakibaei M, Sitte N, Schafer M, Stein C (2004) Subcellular pathways of beta-endorphin synthesis, processing, and release from immunocytes in inflammatory pain. Endocrinology 145:1331-1341.
    Munglani R, Hudspith MJ, Fleming B, Harrisson S, Smith G, Bountra C, Elliot PJ, Birch PJ, Hunt SP (1999) Effect of pre-emptive NMDA antagonist treatment on long-term Fos expression and hyperalgesia in a model of chronic neuropathic pain. Brain Research 822:210-219.
    Muscoli C, Doyle T, Dagostino C, Bryant L, Chen Z, Watkins LR, Ryerse J, Bieberich E, Neumman W, Salvemini D (2010) Counter-regulation of opioid analgesia by glial-derived bioactive sphingolipids. The Journal of Neuroscience : the official journal of the Society for Neuroscience 30:15400-15408.
    Nguyen HX, Galvan MD, Anderson AJ (2008) Characterization of early and terminal complement proteins associated with polymorphonuclear leukocytes in vitro and in vivo after spinal cord injury. J Neuroinflammation 5:26.
    Nilsson A, Moller K, Dahlin L, Lundborg G, Kanje M (2005) Early changes in gene expression in the dorsal root ganglia after transection of the sciatic nerve; effects of amphiregulin and PAI-1 on regeneration. Brain Res Mol Brain Res 136:65-74.
    Papadopoulos CM, Tsai SY, Cheatwood JL, Bollnow MR, Kolb BE, Schwab ME, Kartje GL (2006) Dendritic plasticity in the adult rat following middle cerebral artery occlusion and Nogo-a neutralization. Cereb Cortex 16:529-536.
    Poisbeau P, Patte-Mensah C, Keller AF, Barrot M, Breton JD, Luis-Delgado OE, Freund-Mercier MJ, Mensah-Nyagan AG, Schlichter R (2005) Inflammatory pain upregulates spinal inhibition via endogenous neurosteroid production. The Journal of Neuroscience : the official journal of the Society for Neuroscience 25:11768-11776.
    Pollari E, Savchenko E, Jaronen M, Kanninen K, Malm T, Wojciechowski S, Ahtoniemi T, Goldsteins G, Giniatullina R, Giniatullin R, Koistinaho J, Magga J (2011) Granulocyte colony stimulating factor attenuates inflammation in a mouse model of amyotrophic lateral sclerosis. Journal of Neuroinflammation 8:74.
    Przewlocki R, Przewlocka B (2005) Opioids in neuropathic pain. Current Pharmaceutical Design 11:3013-3025.
    Rittner HL, Brack A, Machelska H, Mousa SA, Bauer M, Schafer M, Stein C (2001) Opioid peptide-expressing leukocytes: identification, recruitment, and simultaneously increasing inhibition of inflammatory pain. Anesthesiology 95:500-508.
    Rittner HL, Brack A, Stein C (2008) The other side of the medal: how chemokines promote analgesia. Neurosci Lett 437:203-208.
    Rittner HL, Labuz D, Schaefer M, Mousa SA, Schulz S, Schafer M, Stein C, Brack A (2006a) Pain control by CXCR2 ligands through Ca2+-regulated release of opioid peptides from polymorphonuclear cells. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 20:2627-2629.
    Rittner HL, Mousa SA, Labuz D, Beschmann K, Schafer M, Stein C, Brack A (2006b) Selective local PMN recruitment by CXCL1 or CXCL2/3 injection does not cause inflammatory pain. Journal of Leukocyte Biology 79:1022-1032.
    Ro LS, Li HY, Huang KF, Chen ST (2004) Territorial and extra-territorial distribution of Fos protein in the lumbar spinal dorsal horn neurons in rats with chronic constriction nerve injuries. Brain Research 1004:177-187.
    Roberts AW (2005) G-CSF: a key regulator of neutrophil production, but that's not all! Growth Factors 23:33-41.
    Rodella L, Rezzani R, Gioia M, Tredici G, Bianchi R (1998) Expression of Fos immunoreactivity in the rat supraspinal regions following noxious visceral stimulation. Brain Research Bulletin 47:357-366.
    Rothwell NJ (1999) Annual review prize lecture cytokines - killers in the brain? The Journal of Physiology 514 ( Pt 1):3-17.
    Rutella S (2007) Granulocyte colony-stimulating factor for the induction of T-cell tolerance. Transplantation 84:S26-30.
    Sager PT, Perlmutter RA, Rosenfeld LE, McPherson CA, Wackers FJ, Batsford WP (1988) Electrophysiologic effects of thrombolytic therapy in patients with a transmural anterior myocardial infarction complicated by left ventricular aneurysm formation. Journal of the American College of Cardiology 12:19-24.
    Satoh J, Onoue H, Arima K, Yamamura T (2005) Nogo-A and nogo receptor expression in demyelinating lesions of multiple sclerosis. Journal of Neuropathology and Experimental Neurology 64:129-138.
    Satoh J, Tabunoki H, Yamamura T, Arima K, Konno H (2007) TROY and LINGO-1 expression in astrocytes and macrophages/microglia in multiple sclerosis lesions. Neuropathology and Applied Neurobiology 33:99-107.
    Schweizerhof M, Stosser S, Kurejova M, Njoo C, Gangadharan V, Agarwal N, Schmelz M, Bali KK, Michalski CW, Brugger S, Dickenson A, Simone DA, Kuner R (2009) Hematopoietic colony-stimulating factors mediate tumor-nerve interactions and bone cancer pain. Nat Med 15:802-807.
    Semerad CL, Poursine-Laurent J, Liu F, Link DC (1999) A role for G-CSF receptor signaling in the regulation of hematopoietic cell function but not lineage commitment or differentiation. Immunity 11:153-161.
    Seymour AB, Andrews EM, Tsai SY, Markus TM, Bollnow MR, Brenneman MM, O'Brien TE, Castro AJ, Schwab ME, Kartje GL (2005) Delayed treatment with monoclonal antibody IN-1 1 week after stroke results in recovery of function and corticorubral plasticity in adult rats. Journal of Cerebral Blood Flow and Metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 25:1366-1375.
    Shamash S, Reichert F, Rotshenker S (2002) The cytokine network of Wallerian degeneration: tumor necrosis factor-alpha, interleukin-1alpha, and interleukin-1beta. The Journal of Neuroscience : the official journal of the Society for Neuroscience 22:3052-3060.
    Solaroglu I, Jadhav V, Zhang JH (2007) Neuroprotective effect of granulocyte-colony stimulating factor. Front Biosci 12:712-724.
    Sotgiu ML, Biella G, Firmi L, Pasqualucci V (1998) Topical axonal transport blocker vincristine prevents nerve injury-induced spinal neuron sensitization in rats. Journal of Neurotrauma 15:1077-1082.
    Stein C, Hassan AH, Przewlocki R, Gramsch C, Peter K, Herz A (1990) Opioids from immunocytes interact with receptors on sensory nerves to inhibit nociception in inflammation. Proc Natl Acad Sci U S A 87:5935-5939.
    Stein C, Schafer M, Machelska H (2003) Attacking pain at its source: new perspectives on opioids. Nat Med 9:1003-1008.
    Stuesse SL, Cruce WL, Lovell JA, McBurney DL, Crisp T (2000) Microglial proliferation in the spinal cord of aged rats with a sciatic nerve injury. Neuroscience Letters 287:121-124.
    Takemoto Y, Wada H, Takatsuka H, Okamoto T, Kakishita E (2000) Mobilization of peripheral blood stem cells by granulocyte-colony stimulating factors: comparison of a standard dose of glycosylated and mutated granulocyte-colony stimulating factor in non-Hodgkin's lymphoma patients following CHOP therapy. Drugs Exp Clin Res 26:1-5.
    Tanga FY, Raghavendra V, DeLeo JA (2004) Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain. Neurochemistry International 45:397-407.
    Tehranian R, Andell-Jonsson S, Beni SM, Yatsiv I, Shohami E, Bartfai T, Lundkvist J, Iverfeldt K (2002) Improved recovery and delayed cytokine induction after closed head injury in mice with central overexpression of the secreted isoform of the interleukin-1 receptor antagonist. Journal of Neurotrauma 19:939-951.
    Tian DS, Liu JL, Xie MJ, Zhan Y, Qu WS, Yu ZY, Tang ZP, Pan DJ, Wang W (2009) Tamoxifen attenuates inflammatory-mediated damage and improves functional outcome after spinal cord injury in rats. Journal of Neurochemistry 109:1658-1667.
    Toulmond S, Rothwell NJ (1995) Interleukin-1 receptor antagonist inhibits neuronal damage caused by fluid percussion injury in the rat. Brain Research 671:261-266.
    Touw IP, van de Geijn GJ (2007) Granulocyte colony-stimulating factor and its receptor in normal myeloid cell development, leukemia and related blood cell disorders. Front Biosci 12:800-815.
    Truong W, Cheng C, Xu QG, Li XQ, Zochodne DW (2003) Mu opioid receptors and analgesia at the site of a peripheral nerve injury. Annals of Neurology 53:366-375.
    Tsai RK, Chang CH, Wang HZ (2008) Neuroprotective effects of recombinant human granulocyte colony-stimulating factor (G-CSF) in neurodegeneration after optic nerve crush in rats. Exp Eye Res 87:242-250.
    Ugolini G, Marinelli S, Covaceuszach S, Cattaneo A, Pavone F (2007) The function neutralizing anti-TrkA antibody MNAC13 reduces inflammatory and neuropathic pain. Proc Natl Acad Sci U S A 104:2985-2990.
    Urdzikova L, Jendelova P, Glogarova K, Burian M, Hajek M, Sykova E (2006) Transplantation of bone marrow stem cells as well as mobilization by granulocyte-colony stimulating factor promotes recovery after spinal cord injury in rats. Journal of Neurotrauma 23:1379-1391.
    Venkatesh K, Chivatakarn O, Lee H, Joshi PS, Kantor DB, Newman BA, Mage R, Rader C, Giger RJ (2005) The Nogo-66 receptor homolog NgR2 is a sialic acid-dependent receptor selective for myelin-associated glycoprotein. The Journal of Neuroscience : the official journal of the Society for Neuroscience 25:808-822.
    Wang F, Shen X, Guo X, Peng Y, Liu Y, Xu S, Yang J (2010) Spinal macrophage migration inhibitory factor contributes to the pathogenesis of inflammatory hyperalgesia in rats. Pain 148:275-283.
    Watkins LR, Maier SF (2002) Beyond neurons: evidence that immune and glial cells contribute to pathological pain states. Physiological Reviews 82:981-1011.
    Whalen MJ, Carlos TM, Wisniewski SR, Clark RS, Mellick JA, Marion DW, Kochanek PM (2000) Effect of neutropenia and granulocyte colony stimulating factor-induced neutrophilia on blood-brain barrier permeability and brain edema after traumatic brain injury in rats. Critical Care Medicine 28:3710-3717.
    White FA, Jung H, Miller RJ (2007) Chemokines and the pathophysiology of neuropathic pain. Proc Natl Acad Sci U S A 104:20151-20158.
    Wu J, Fang L, Lin Q, Willis WD (2001) Nitric oxide synthase in spinal cord central sensitization following intradermal injection of capsaicin. Pain 94:47-58.
    Yan J, Zhou X, Guo JJ, Mao L, Wang YJ, Sun J, Sun LX, Zhang LY, Zhou XF, Liao H (2012) Nogo-66 inhibits adhesion and migration of microglia via GTPase Rho pathway in vitro. Journal of Neurochemistry 120:721-731.
    Yan Z, Stapleton PP, Freeman TA, Fuortes M, Daly JM (2004) Enhanced expression of cyclooxygenase-2 and prostaglandin E2 in response to endotoxin after trauma is dependent on MAPK and NF-kappaB mechanisms. Cellular Immunology 232:116-126.
    Zhang RX, Liu B, Wang L, Ren K, Qiao JT, Berman BM, Lao L (2005) Spinal glial activation in a new rat model of bone cancer pain produced by prostate cancer cell inoculation of the tibia. Pain 118:125-136.
    Zhang XY, Zhou Y, Liu WQ, Zhao L, Li SQ (2007) [Effects of antisense oligonucleotide to nuclear factor-kappaB on the development of bleomycin-induced pulmonary fibrosis and IL-4 expression therein: experiment with mice]. Zhonghua yi xue za zhi 87:195-199.
    Zhang YQ, Guo N, Peng G, Wang X, Han M, Raincrow J, Chiu CH, Coolen LM, Wenthold RJ, Zhao ZQ, Jing N, Yu L (2009) Role of SIP30 in the development and maintenance of peripheral nerve injury-induced neuropathic pain. Pain 146:130-140.
    Zhao P, Waxman SG, Hains BC (2007) Extracellular signal-regulated kinase-regulated microglia-neuron signaling by prostaglandin E2 contributes to pain after spinal cord injury. The Journal of Neuroscience : the official journal of the Society for Neuroscience 27:2357-2368.
    Zhuo M, Wu G, Wu LJ (2011) Neuronal and microglial mechanisms of neuropathic pain. Molecular Brain 4:31.

    下載圖示
    QR CODE