INTERLEUKIN 6 RELEASE FROM t98g HUMAN GLIAL CELL LINE AS a PREDICTIVE MARKER FOR CHRONIC PAIN, AND THE CHARACTERIZATION OF SUBSTANCE(S) INVOLVED IN PAIN

136 600 0
INTERLEUKIN 6 RELEASE FROM t98g HUMAN GLIAL CELL LINE AS a PREDICTIVE MARKER FOR CHRONIC PAIN, AND THE CHARACTERIZATION OF SUBSTANCE(S) INVOLVED IN PAIN

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

INTERLEUKIN-6 RELEASE FROM T98G HUMAN GLIAL CELL LINE AS A PREDICTIVE MARKER FOR CHRONIC PAIN, AND THE CHARACTERIZATION OF SUBSTANCE(S) INVOLVED IN CHRONIC PAIN TAY SUAN ANNABEL B.Sc (Hons.), NUS A THESIS SUBMITTED FOR THE DEGREE OF MASTER OF SCIENCE DEPARTMENT OF ANAESTHESIA NATIONAL UNIVERSITY OF SINGAPORE 2013 i Acknowledgements I would like to express my sincere gratitude to my supervisor A/P Low Chian Ming, for his support, advice and constant encouragement throughout my research career at the National University of Singapore Throughout the course of my project, I have not only learned many laboratory techniques, but also learned many invaluable life skills that will benefit me for life I would also like to express my gratitude to Prof Shinro Tachibana, for his guidance and support throughout the course of my project Without his directions, this work would not have come to fruition My heartfelt thanks are also due to A/P Liu Hern Choon Eugene and Prof Lee Tat Leang, for supporting my work constantly, for their helpful advice when things did not go well, and for helping me procure the valuable samples from National University Hospital for the project Thank you for your supervision, guidance and support throughout my study My sincere thanks also goes out to Prof Toshiaki Minami for helping me procure the precious samples from Osaka Medical University Hospital for my research work I would also like to express my heartfelt thanks to Mdm Li Chunmei for her constant support, advice and encouragement Thank you for all your help with my HPLC work and other technical support My sincere thanks go out to Ms Wang Anni for her assistance in western blot and for brightening up my days as a researcher Thank you both so much for all the fun and laughter you have brought to my days in the laboratory I am also very grateful to Ms Jeyapriya Raja Sundaram for her help and guidance in primary astrocyte culture and immunocytochemistry, and for her kind advice throughout my study My sincere ii thanks are also due to Mdm Karen Ho Ban Shian and Mrs Mariam Mathew for their administrative support and assistance Lastly, I am grateful to my family and friends for their understanding, endless support and encouragement throughout the course of my research work iii Table of Contents Acknowledgements ii Summary ix List of Tables xi List of Figures xii List of Abbreviations xiv List of Publications xvii List of Conference Papers xvii Chapter Introduction 1.1 Chronic Pain 1.1.1 Epidemiology of Chronic Pain 1.1.2 Pathophysiology of Chronic Pain 1.1.2.1 Role of Glial Cells in Chronic Pain 1.1.2.1.1 T98G Cell Line as an in vitro Astrocytic Model 1.1.3 Treatment Strategies for Chronic Pain and their Challenges 1.1.4 Conditions related to Chronic Pain 11 1.1.4.1 Post-Herpetic Neuralgia (PHN) 11 1.1.4.2 Osteoarthritis 12 1.2 Neurotransmitters/Neuromodulators in Chronic Pain 1.2.1 Small Molecule Neurotransmitters 14 14 1.2.1.1 Amino Acids 14 1.2.1.2 Prostaglandins 16 1.2.2 Neuropeptides 16 1.2.2.1 Substance P 17 iv 1.2.2.2 Nociceptin/orphanin FQ and Nocistatin 1.2.3 Pro-inflammatory Cytokines 17 18 1.2.3.1 Tumour Necrosis Factor-α (TNF-α) 19 1.2.3.2 Interleukin-1β (IL-1β) 21 1.2.3.3 Interleukin-6 (IL-6) 22 1.3 Importance of Cerebrospinal Fluid (CSF) in Chronic Pain Research 1.3.1 Biological Markers in the CSF 1.4 Aim and Scope of Study Chapter 23 24 26 Release of Pro-inflammatory Cytokines in T98G Cells Upon Exposure to CSF of PHN Patients 2.1 Objectives of Chapter 29 2.2 Materials and Methods 31 2.2.1 Materials 31 2.2.2 CSF Samples 32 2.2.3 Cell Culture 33 2.2.4 Measurement of TNF-α, IL-1β and IL-6 Release 34 2.2.5 Measurement of Dexamethasone in CSF 35 2.2.6 Statistical Analyses 36 2.3 Results 37 2.3.1 TNF-α, IL-1β and IL-6 Releasing Activity in T98G Cells Upon Exposure to CSF of PHN Patients 37 2.3.2 Comparison of IL-6 Releasing Activity Between CSF of Different PHN Treatment Groups 39 2.3.3 Effect of in vitro Steroid on IL-6 Releasing Activity 2.4 Discussion 41 44 v Chapter Comparison of IL-6 Releasing Activity between CSF of Chronic Pain Patients and Pain-free Patients 3.1 Objectives of Chapter 49 3.2 Materials and Methods 50 3.2.1 Materials 50 3.2.2 CSF Samples 50 3.2.3 Culture of Primary Astrocytes 51 3.2.4 Immunocytochemistry 52 3.2.5 Measurement of IL-6 in T98G and Primary Astrocyte Cell Culture 53 3.2.6 Statistical Analyses 53 3.3 Results 54 3.3.1 Comparison between PHN Therapy Effective, Therapy Ineffective and Control Group 54 3.3.2 Comparison between Osteoarthritis and Control Group 56 3.3.3 IL-6 Release from Primary Astrocytes Upon Exposure to CSF of Chronic Pain Patients 57 3.4 Discussion Chapter 59 Purification of Protein-like Compounds from CSF of Chronic Pain Patients 4.1 Objectives of Chapter 64 4.2 Materials and Methods 65 4.2.1 Materials 65 4.2.2 CSF Samples 65 4.2.3 Preliminary Experiments on CSF 66 4.2.3.1 Separation of CSF by Molecular Weight vi 66 4.2.3.2 Treatment of CSF with Pronase 66 4.2.3.3 Measurement of IL-6 in T98G Cell Culture 67 4.2.4 Separation of CSF into Different Fractions 67 4.2.4.1 CSF Fractionation using HPLC 68 4.2.4.2 Measurement of IL-6 in T98G Cell Culture 70 4.2.5 IL-6 Release from T98G Cells When Exposed to Lignocaine and Albumin 70 4.2.5 Statistical Analyses 71 4.3 Results 72 4.3.1 CSF Fraction >10 kDa Molecular Weight Triggered IL-6 Release in T98G Cells 72 4.3.2 Pronase Attenuated IL-6 Release in T98G Cells 74 4.3.3 Separation of CSF by HPLC 75 4.3.3.1 HPLC on Pooled CSF 75 4.3.3.2 HPLC on Pooled Active Fractions 77 4.3.4 Comparison between Chromatograms Obtained from HPLC of Osteoarthritis CSF and Control CSF 4.3.5 Effects of Lignocaine and Albumin on IL-6 Release 4.4 Discussion Chapter 80 81 83 Signaling Mechanism of Chronic Pain in the T98G Cell System 5.1 Objectives of Chapter 89 5.2 Materials and Methods 90 5.2.1 Materials 90 5.2.2 CSF Samples 90 5.2.3 Cell Culture and NF-κB Inhibition 91 vii 5.2.4 Cell Fractionation and Cell Lysis 91 5.2.5 Western Blot 93 5.2.6 Statistical Analyses 93 5.3 Results 95 5.3.1 Inhibition of NF-κB led to Reduced IL-6 Release in T98G Cells 95 5.3.2 NF-κB Activation in T98G Cells Upon Exposure to CSF of Chronic Pain Patients 97 5.4 Discussion Chapter 99 Conclusion and Future Directions 6.1 Conclusion 104 6.2 Limitations of Study 106 6.3 Future Directions 107 References 109 viii Summary Besides neurons, the central nervous system (CNS) consists of glial cells, which are mainly microglia and astrocytes Chronic pain is classically viewed as being mediated solely by neurons, but there is mounting evidence that glial cells also play a part Glial cells, responding to stimulation by neurotransmitters and peptides, are activated and release pain-enhancing substances like proinflammatory cytokines These cytokines have been shown to play a role in enhancing pain by their actions in the spinal cord This research work focuses firstly on investigating pro-inflammatory cytokine release in a cell culture system as a potential marker for chronic pain Two conditions related to chronic pain were studied: post-herpetic neuralgia (PHN) and osteroarthritis Cerebrospinal fluid (CSF) from patients suffering from either condition was used to trigger astrocytic cell line T98G cultures, and subsequent pro-inflammatory cytokine release was measured by enzyme-linked immunosorbent assay (ELISA) IL-6 release in the chronic pain patient groups was found to be significantly higher compared to pain-free controls, as well as in the PHN patient group whose steroid treatment was ineffective compared to those whose treatment was effective CSF samples collected before steroid treatment also triggered higher IL-6 release than after treatment samples These in vitro tests provide an objective evaluation on the extent of chronic pain as well as the efficacy of steroid therapy ix 6.1 Conclusion Chronic pain is a major healthcare problem due to its negative impact on work performance and daily activities Effective therapeutics for chronic pain requires targeting the cellular mechanisms that individually or collectively produce pain, instead of targeting superficial symptoms Hence, studying the mechanisms underlying chronic pain and the search for potential targets for chronic pain treatment have become top priority in pain research in recent years The importance of biological markers in chronic pain research is apparent, be it in assessing the degree of chronic pain, in enabling the early diagnosis of painrelated diseases, or in predicting the outcome of pain treatment There are no reports on in vitro markers that may aid in chronic pain research at present In this pursuit in understanding the complex mechanisms of chronic pain, this study focused on establishing an in vitro biological marker that may aid in the management of chronic pain, and also on isolation and identification of substances involved in chronic pain The first objective of the study was to investigate pro-inflammatory cytokine release in a T98G cell culture system as a potential marker for chronic pain IL-6 release from T98G cells was found to be elevated upon exposure to CSF of chronic pain patients as compared to CSF of pain-free controls IL-6 release was also elevated upon exposure to CSF collected before steroid therapy as well as upon exposure to CSF of patients whose steroid therapy was ineffective IL-6 release upon exposure to CSF of patients whose therapy was effective was attenuated completely when in vitro steroid was added to the cell culture All 104 these results showed the effectiveness of IL-6 release in the T98G cell culture system upon addition of CSF as in vitro biological marker for chronic pain The second objective of the study was to fractionate and isolate painrelated substances in the CSF and to establish the signaling pathway involved in chronic pain CSF from chronic osteoarthritic pain patients was fractionated by a two-step HPLC procedure, using the previously established cell culture system to analyze the IL-6 releasing ability of each peak Three peaks that could trigger IL6 release in T98G cells were isolated and collected at the end of the procedures The HPLC chromatogram of chronic pain CSF was compared against that of painfree control CSF and the peaks that showed differences were also collected We have effectively isolated fractions of chronic osteoarthritic pain CSF that contain protein-like substances that could contribute to chronic pain In addition, in investigating the possible signaling mechanism that leads to the IL-6 release, it was found that exposure to chronic osteoarthritic pain CSF led to NF-κB activation in T98G cells and this activation acts upstream of IL-6 In summary, the present results of the study demonstrate another perspective in chronic pain research Utilizing CSF of chronic pain patients, IL-6 have been successfully established as an in vitro biological marker that can provide an objective evaluation of the extent of chronic pain, which is an advantage over subjective methods such as pain score measurements, and also to predict an individual’s response to i.t steroid therapy The isolation of fractions from chronic osteoarthritic pain CSF that can trigger IL-6 release paves a way for the identification and characterization of pain-related substances in the CSF 105 These collected fractions, after analysis by mass spectrometry, would provide highly useful and detailed information on the protein-like substances in the CSF that contribute to chronic pain Establishing that CSF of chronic pain patients trigger NF-κB activation in T98G cells upstream of IL-6 is the first step towards elucidating the mechanisms behind IL-6 release in this T98G cell system NF-κB could be yet another useful in vitro biological marker for the objective assessment of chronic pain 6.2 Limitations of Study The study involves in vitro work using T98G cells as an astrocytic model The disadvantage of in vitro studies is that it is difficult to extrapolate the results achieved from in vitro studies to the actual physiology of humans The results obtained using T98G cells could not be replicated using primary astrocytes As such, the IL-6 release from T98G cells acts uniquely as a read-out, providing an unbiased assessment of the extent of chronic pain in patients Another limitation of our study was that due to difficulties in obtaining more CSF samples, we were only able to focus on two conditions related to chronic pain: PHN and osteoarthritis In addition, due to the lack of more PHN CSF samples, isolation of pain-related protein-like substances using HPLC could only be carried out on osteoarthritis CSF samples Encompassing more chronic pain conditions in our study would widen the scope of our work Comparisons can be made between various pain conditions, thereby establishing a more complete 106 picture of the relationship between substances in the CSF and the perception of chronic pain due to different conditions 6.3 Future Directions The separation and purification of chronic pain patients’ CSF into different fractions is just the first step in the characterization of substances involved in chronic pain More in depth studies have to be performed on these purified fractions MALDI-TOF mass spectrometry would be the next step to determine the molecular weight of the peaks, and the peaks would be subsequently sequenced to identify and characterize the protein-like substances in the CSF that play a role in chronic pain Subsequently, these substances could be isolated and animal bioactivity studies carried out to determine the pain perception mechanisms of these substances Further work should also be carried out on the signaling mechanism of IL6 release Identification of NF-κB activation as upstream of IL-6 release is only the first step in unraveling the signaling mechanism of chronic pain in this T98G cell system Other substances known to be involved in IL-6 activation and chronic pain mechanisms should also be analyzed Due to the relatively invasive nature of obtaining CSF from patients as well as the limited amount (2 mL) that can be withdrawn at any one time, it would be beneficial if a substitute that is more easily obtained can be used in place of CSF It was demonstrated that the concentration of 13 amino acids in CSF was 107 directly related to the plasma concentration in normal individuals (McGale et al., 1977) Levels of serum and CSF glucose were also positively correlated in children (Nigrovic et al., 2012) It would be worthwhile to investigate the correlation between chronic pain patients’ serum and CSF on their ability to trigger IL-6 release in T98G cells If there is a correlation, this method would potentially serve as a valuable analytical tool in the assessment of chronic pain, and more chronic pain conditions could be studied as well, as human serum samples are more easily and more conveniently collected 108 References W.E Ackerman IV, T.L.S Summerfield, D.D Vandre, J.M Robinson, D.A Kniss, Nuclear Factor-Kappa B regulates inducible prostaglandin E synthase expression in human amnion mesenchymal cells, Biol Reprod 78 (2008) 68–76 H Alaranta, M Hurme, K Lahtela, M.T Hyyppa, Prolactin and cortisol in cerebrospinal fluid: sex-related associations with clinical and psychological characteristics of patients with low back pain, Psychoneuroendocrinol 8:3 (1983) 333-341 G.M Alexander, M.A van Rijn, J.J van Hilten, M.J Perreault, R.J Schwartzman, Changes in cerebrospinal fluid levels of pro-inflammatory cytokines in CRPS, Pain 116 (2005) 213-219 G.M Alexander, M.J Perreault, E.R Reichenberger, R.J Schwartzman, Changes in immune and glial markers in the CSF of patients with Complex Regional Pain Syndrome, Brain Behav Immun 21:5 (2007) 668-676 J.L Arruda, R.W Colburn, A.J Rickman, M.D Rutkowski, J.A DeLeo, Increase of interleukin-6 mRNA in the spinal cord following peripheral nerve injury in the rat: potential role of IL-6 in neuropathic pain, Mol Brain Res 62 (1998) 228-235 M Backonja, C.L Coe, D.A Muller, K Schell, Altered cytokine levels in the blood and cerebrospinal fluid of chronic pain patients, J Neuroimmunol 195 (2008) 157-163 L Bazzichi, A Rossi, G Massimetti, G Giannaccini, T Giuliano, F De Feo, A Ciapparelli, L Dell’Osso, S Bombardieri, Cytokine patterns in fibromyalgia and their correlation with clinical manifestations, Clin Exp Rheumatol 25 (2007) 225-230 J.S Bennett, A Daugherty, D Herrington, P Greenland, H Roberts, K.A Taubert, The use of nonsteroidal anti-inflammatory drugs (NSAIDs), Circulation 111(2005) 1713-1716 H Breivik, B Collett, V Ventafridda, R Cohen, D Gallacher, Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment, Eur J Pain 10:4 (2006) 287-333 Y.Q Cao, P.W Mantyh, E.J Carlson, A.-M Gillespie, C.J Epstein, A.I Basbaum, Primary afferent tachykinins are required to experience moderate to intense pain, Nature 392 (1998), 390-394 H.N Chan, B.Y Ng, Use of antidepressants in the treatment of chronic pain, Ann Acad Med Singapore 38 (2009) 974-979 109 B.A Chizh, Novel approaches to targeting glutamate receptors for the treatment of chronic pain: review article, Amino Acids 23 (2002) 167-176 F Cunha, S Poole, B Lorenzetti, S Ferreira, The pivotal role of tumor necrosis factor alpha in the development of inflammatory hyperalgesia, Br J Pharmacol 107 (1992) 660-664 P Conaghan, Osteoarthritis: national clinical guideline for care and management in adults, London: Royal College of Physicians (2008) A.C de Joannon, F Mancini, C Landolfi, L Soldo, A Leta, A Ruggieri, G Mangano, L Polenzani, M Pinza, C Milanese, Adenosine triphosphate affects interleukin-1β release by T98G glioblastoma cells through a purinoceptorindependent mechanism, Neurosci Lett 285:3 (2000) 218-222 R.F De Jongh, K.C Vissers, T.F Meert, L.H.D.J Booij, C.S De Deyne, R.J Heylen, The role of interleukin-6 in nociception and pain, Anesth Analg 96 (2003) 1099-1103 M de Mos, A Laferriere, M Millecamps, M Pilkington, M.C.J.M Sturkenboom, F.J.P.M Huygen, T.J Coderre, Role of NF-κB in an animal model of Complex Regional Pain Syndrome–type I (CRPS-I), J Pain 10 (2009) 1161-1169 J.A DeLeo, R.W Colburn, A.J Rickman, Cytokine and growth factor immunohistochemical spinal profiles in two animal models of mononeuropathy, Brain Res 759 (1997) 50-57 C De Felipe, J.F Herrero, J.A O'Brien, J.A Palmer, C.A Doyle, A.J Smith, J.M Laird, C Belmonte, F Cervero, S.P Hunt, Altered nociception, analgesia and aggression in mice lacking the receptor for substance P Nature 392 (1998) 394– 397 S.A Doppenschmitt, B Scheidel, F Harrison, J.P Surmann, Simultaneous determination of prednisolone, prednisolone acetate and hydrocortisone in human serum by high-performance liquid chromatography, J Chromatography 674 (1995) 237-246 G.P Dureja, H Usmani, M Khan, M Tahseen, A Jamal, Efficacy of intrathecal midazolam with or without epidural methylprednisolone for management of postherpetic neuralgia involving lumbosacral dermatomes, Pain Physician 13 (2010) 213-221 R.H Dworkin, A.B O’Connor, M Backonja, J.T Farrar, N.B Finnerup, T.S Jensen, E.A Kalso, J.D Loeser, C Miaskowski, T.J Nurmikko, R.K Portenoy, A.S.C Rice, B.R Stacey, R.-D Treede, D.C Turk, M.S Wallace, Pharmacologic management of neuropathic pain: evidence-based recommendations, Pain 132 (2007) 237-251 110 M.J Eaton, M.A Martinez, S Karmally, A single intrathecal injection of GABA permanently reverses neuropathic pain after nerve injury, Brain Res 835 (1999) 334-339 D.T Felson, Epidemiology of the rheumatic diseases, In: Arthritis and allied conditions: a textbook of rheumatology, 13th Edition, Ed: W.J Koopman, Williams & Wilkins, Baltimore (1997) S Ferreira, B Lorenzetti, A Bristow, S Poole, Interleukin-1β as a potent hyperalgesic agent antagonized by a tripeptide analogue, Nature 334 (1988) 698700 E.S Fu, Y P Zhang, J Sagen, Z.Q Yang, J.R Bethea, Transgenic glial nuclear factor-κ B inhibition decreases formalin pain in mice, Neuroreport 18 (2007) 713717 R.A Gadient, U.H Otten, Interleukin-6: a molecule with both beneficial and destructive potentials, Progess Neurobiol 52 (1997) 379-390 Y.-J Gao, R.-R Ji, Targeting astrocyte signaling for chronic pain, Neurotherapeutics 7:4 (2010) 482-493 P Gasque, P Chan, C Mauger, M.T Schouft, S Singhrao, M.P Dierich, B.P Morgan, M Fontaine, Identification and characterization of complement C3 receptors on human astrocytes, J Immunol 156:6 (1996) 2247-2255 A George, C Schmidt, A Weishaupt, K.V Toyka, C Sommer, Serial determination of tumor necrosis factor-α content in rat sciatic nerve after chronic constriction injury, Exp Neurol 160 (1999) 124-132 S.R Goldring, M.B Goldring, Clinical aspects, pathology and pathophysiology of osteoarthritis, J Musculoskelet Neuronal Interact 6:4 (2006) 376-378 C.R Goucke, The management of persistent pain, Med J Aust 178 (2003) 444447 W Guo, H Wang, M Watanabe, K Shimizu, S Zou, S.C LaGraize, F Wei, R Dubner, K Ren, Glial-cytokine-neuronal interactions underlying the mechanisms of persistent pain, J Neurosci 27 (2007) 6006-6018 A Gur, M Karakoc, K Nas, C Remzi, A Denli, J Sarac, Cytokines and depression in cases with fibromyalgia, J Rheumatol 29 (2002) 358-361 M.M Halassa, T Fellin, H Takano, J.H Dong, P.G Haydon, Synaptic islands defined by the territory of a single astrocyte, J Neurosci 27 (2007) 6473-6477 A Hald, S Nedegaard, R.R Hansen, M Ding, A.M Heegaard, Differential activation of spinal cord glial cells in murine models of neuropathic and cancer pain, Eur J Pain 13 (2009) 138-145 111 E Hanson, L Ronnback, Astrocytic receptors and second messenger systems, Adv Mol Cell Biol 31 (2004) 475-501 H Hashizume, J.A DeLeo, R.W Colburn, J.N Weinstein, Spinal glial activation and cytokine expression after lumbar root injury in the rat, Spine 25 (2000) 12061217 G Heine, H.D Zucht, M.U Schuhmann, K Burger, M Jurgens, M Zumkeller, C.G Schneekloth, H Hampel, P Schulz-Knappe, H Selle, High-resolution peptide mapping of cerebrospinal fluid: a novel concept for diagnosis and research in central nervous system diseases, J Chromatogr B 782 (2002) 353-61 K.Y Ho, C Siau, Chronic pain management: therapy, drugs and needles, Ann Acad Med Singapore 38 (2009) 929-930 M Holtta, H Zetterberg, E Mirgorodskaya, N Mattsson, K Blennow, J Gobom, Peptidome analysis of cerebrospinal fluid by LC-MALDI MS, PLoS ONE 7:8 (2012) e42555 R.G Holzheimer, W Steinmetz, Local and systemic concentrations of pro- and anti-inflammatory cytokines in human wounds, Eur J Med Res 18 (2000) 347355 T Hoshina, Y Yamaguchi, S Ohga, R Kira, M Ishimura, H Takada, T Tanaka, T Hara, Sjogren’s syndrome-associated meningoencephalomyelitis: cerebrospinal fluid cytokine levels and therapeutic utility of tacrolimus, J Neurol Sci 267 (2008) 182-186 A.F Huhmer, R.G Biringer, H Amato, A.N Fonteh, M.G Harrington, Protein analysis in human cerebrospinal fluid: physiological aspects, current progress and future challenges, Dis Markers 22 (2006) 3–26 F Iannone, G Lapadula, The pathophysiology of osteoarthritis, Aging Clin Exp Res 15:5 (2003) 364-372 T Ichiyama, H Isumi, T Yoshitomi, M Nishikawa, T Matsubara, S Furukawa, NF-kappaB activation in cerebrospinal fluid cells from patients with meningitis, Neurol Res 24:7 (2002) 709-712 Y Ito, M Yamamoto, M Li, M Doyu, F Tanaka, T Mutch, T Mitsuma, G Sobue, Differential temporal expression of mRNAs for ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), interleukin-6 (IL-6), and their receptors (CNTFR alpha, LIFR beta, IL-6R alpha and gp130) in injured peripheral nerves, Brain Res 793 (1998) 321-327 C.E Johanson, J.A Duncan 3rd, P.M Klinge, T Brinker, E.G Stopa, G.D Silverberg, Multiplicity of cerebrospinal fluid functions: New challenges in health and disease, Cerebrospinal Fluid Res (2008) 10 112 T Joseph, T.L Lee, C Li, C Siau, Y Nishiuchi, T Kimura, S Tachibana, Levels of neuropeptides nocistatin, nociception/orphanin FQ and their precursor protein in a rat neuropathic pain model, Peptides 28 (2007) 1433-1440 R Kolhekar, S.T Meller, G.F Gebhart, N-methyl-D-aspartate receptor-mediated changes in thermal nociception: allosteric modulation at glycine and polyamine recognition sites, Neuroscience 63 (1994) 925-936 N Kotani, T Kushikata, H Hashimoto, F Kimura, M Muraoka, M Yodono, M Asai, A Matsuki, Intrathecal methylprednisolone for intractable postherpetic neuralgia, New Eng J Med 343 (2000) 1514-1519 N Kotani, R Kudo, Y Sakurai, D Sawamura, D.I Sessler, H Okada, H Nakayama, T Yamagata, M Yasujima, A Matsuki, Cerebrospinal fluid interleukin-8 concentrations and the subsequent development of postherpetic neuralgia, Amer J Med 116 (2004) 318-323 B.K Kwon, A.M Stammers, L.M Belanger, A Bernardo, D Chan, C.M Bishop, G.P Slobogean, H Zhang, H Umedaly, M Griffin, J Street, M.C Boyd, S.J Paquette, C.G Fisher, M.F Dvorak, Cerebrospinal fluid inflammatory cytokines and biomarkers of injury severity in acute human spinal cord injury, J Neurotrauma 27 (2010) 669-682 K.M Lee, S.M Jeon, H.J Cho, Tumor necrosis factor receptor induces interleukin-6 upregulation through NF-kappaB in a rat neuropathic pain model, Eur J Pain 13 (2009) 794-806 H.L Lee, K.M Lee, S.J Son, S.H Hwang, H.J Cho, Temporal expression of cytokines and their receptor mRNAs in a neuropathic pain model, Neuroreport 15 (2004) 2807-2811 S.S Leopold, Minimally invasive total knee arthroplasty for osteoarthritis, N Engl J Med 360:17 (2009) 1749-1758 T.A Libermann, D Baltimore, Activation of interleukin-6 gene expression through the NF-κB transcription factor, Mol Cell Biol 10:5 (1990) 2327-2334 C Lindh, Z Liu, S Lyrenas, G Ordeberg, F Nyberg, Elevated cerebrospinal fluid substance P-like immunoreactivity in patients with painful osteoarthritis, but not in patients with rhizopatic pain from a herniated lumbar disc, Scand J Rheumatol 26:6 (1997) 468-472 E.A Lingard, C.B Sledge, I.D Learmonth, Patient expectations regarding total knee arthroplasty: differences among the United States, United Kingdom, and Australia, J Bone Joint Surg Am 88 (2006) 1201-1207 Z Liu, M Welin, B Bragee, F Nyberg, A high-recovery procedure for quantitative analysis of substance P and opioid peptides in human cerebrospinal fluid, Peptides 21 (2000) 853-860 113 J Lu, T Katano, W Nishimura, S Fujiwara, S Miyazaki, I Okasaki, K Aritake, Y Urade, T Minami, S Ito, Proteomic analysis of cerebrospinal fluid before and after intrathecal injection of steroid into patients with postherpetic pain, Proteomics 12 (2012) 1–8 E.H McGale, I.F Pye, C Stonier, E.C Hutchinson, G.M Aber, Studies of the inter-relationship between cerebrospinal fluid and plasma amino acid concentrations in normal individuals, J Neurochem 29 (1977) 291-297 A Meunier, A Latremoliere, E Dominguez, A Mauborgne, S Philippe, M Hamon, J Mallet, J.J Benoliel, M Pohl, Lentiviral-mediated targeted NF-κB blockade in dorsal spinal cord glia attenuates sciatic nerve injury-induced neuropathic pain in the rat, Mol Ther 15 (2007) 687-697 E.D Milligan, K.A O’Connor, K.T Nguyen, C.B Armstrong, C Twining, R.P Gaykema, A Holguin, D Martin, S.F Maier, L.R Watkins, Intrathecal HIV-1 envelope glycoprotein gp120 induces enhanced pain states mediated by spinal cord proinflammatory cytokines, J Neurosci 21 (2001) 2808-2819 K Miyoshi, K Obata, T Kondo, H Okamura, K Noguchi, Interleukin-18mediated microglia/astrocyte interaction in the spinal cord enhances neuropathic pain processing after nerve injury, J Neurosci 28 (2008) 12775-12787 S.J Mnich, A,W Veenhuizen, J.B Monahan, K.C.F Sheehan, K.R Lynch, P.C Isakson, J.P Portanova, Characterization of a monoclonal antibody that neutralizes the activity of prostaglandin E2, J Immunol 155 (1995) 4437-4444 D Mukherjee, S.E Nissen, E.J Topol, Risk of cardiovascular events associated with selective COX-2 inhibitors, JAMA 286 (2001) 954-959 E Niederberger, A Schmidtko, W Gao, H Kuhlein, C Ehnert, G Geisslinger, Impaired acute and inflammatory nociception in mice lacking the p50 subunit of NF-κB, Eur J Pharmacol 559 (2007) 55-60 L.E Nigrovic, A.A Kimia, S.S Shah, M.I Neuman, Relationship between cerebrospinal fluid glucose and serum glucose, N Engl J Med 366 (2012) 576578 J.G Norris, L.-P Tang, S.M Sparacio, E.N Benveniste, Signal transduction pathways mediating astrocyte IL-6 induction by IL-1β and tumor necrosis factorα, J Immunol 152 (1994) 841-850 O Obreja, P.K Rathee, K.S Lips, C Distler, M Kress, IL-1β potentiates heatactivated currents in rat sensory neurons: involvement of IL-1RI, tyrosine kinase, and protein kinase C, FASEB J 16 (2002) 1497-1503 T Oka, S Aou, T Hori, Intracerebroventricular injection of interleukin-1β induces hyperalgesia in rats, Brain Res 624 (1993) 61-68 114 T Oka, S Aou, T Hori, Intracerebroventricular injection of interleukin-1β enhances nociceptive neuronal responses of the trigeminal nucleus caudalis in rats, Brain Res 656 (1994) 236-244 T Oka, K Oka, M Hosoi, T Hori, Intracerebroventricular injection of interleukin-6 induces thermal hyperalgesia in rats, Brain Res 692 (1995) 123-128 W Opstelten, A.J.M van Wijck, R.J Stolker, Interventions to prevent postherpetic neuralgia: cutaneous and percutaneous techniques, Pain 107 (2004) 202-206 P Palmqvist, P Lundberg, I Lundgren, L Hanstrom, U.H Lerner, IL-1beta and TNF-alpha regulate IL-6-type cytokines in gingival fibroblasts, J Dent Res 87:6 (2008) 558-563 W Raffaeli, B.G.S Dekel, D Landuzzi, A Caminiti, D Righetti, M Balestri, F Montanari, P Romualdi, S Candeletti, Nociceptin levels in the cerebrospinal fluid of chronic pain patients with or without intrathecal administration of morphine, J Pain Symptom Manage 32:4 (2006) 372–377 W Ren, Y Liu, L Zhou, W Li, Y Zhong, R Pang, W Xin, X Wei, J Wang, H Zhu, C Wu, Z Qin, G Liu, X Liu, Peripheral nerve injury leads to working memory deficits and dysfunction of the hippocampus by upregulation of TNF-α in rodents, Neuropsychopharm 36 (2011) 979–992 K Roos, Principles of neurologic infectious diseases, McGraw-Hill, New York (2005) T Rozen, S.Z Swidan, Elevation of CSF tumor necrosis factor α levels in new daily persistent headache and treatment refractory chronic migraine, Headache 47 (2007) 1050-1055 A Saravana Kumar, R Gandhimathi, Autopharmacology: a review, Intl J Pharmacol Res (2012) 88-108 M Schafers, D.H Lee, D Brors, T.L Yaksh, L.S Sorkin, Increased sensitivity of injured and adjacent uninjured rat primary sensory neurons to exogenous tumor necrosis factor-α after spinal nerve ligation, J Neurosci 23 (2003) 3028-3038 M Schafers, C.I Svensson, C Sommer, L.S Sorkin, Tumor necrosis factor-α induces mechanical allodynia after spinal nerve ligation by activation of p38 MAPK in primary sensory neurons, J Neurosci 23 (2003) 2517-2521 J Scholz, C.J Woolf, Can we conquer pain? Nat Neurosci Suppl (2002) 10621067 J Scholz, C.J Woolf, The neuropathic pain triad: neurons, immune cells and glia, Nat Neurosci 10:11 (2007) 1361-1368 115 P Schulz-Knappe, H.D Zucht, G Heine, M Jurgens, R Hess, M Schrader, Peptidomics: the comprehensive analysis of peptides in complex biological mixtures, Comb Chem High Throughput Screen (2001) 207-217 D.A Seehusen, M.M Reeves, D.A Fomin, CSF analysis, Am Fam Physician 68:6 (2003) 1103–1108 R Sethuraman, M.G Krishnamoorthy, T.L Lee, E.H Liu, S Chiang, W Nishimura, M Sakai, T Minami, S Tachibana, Simultaneous analysis of D- and L-serine in cerebrospinal fluid by use of HPLC, Clin Chem 53:8 (2007) 14891494 B.L Sng, S.A Schug, The role of opioids in managing chronic non-cancer pain, Ann Acad Med Singapore 38 (2009) 960-966 C Sommer, S Petrausch, T Lindenlaub, K.V Toyka, Neutralizing antibodies to interleukin 1-receptor reduce pain associated behavior in mice with experimental neuropathy, Neurosci Lett 270 (1999) 25-28 C Sommer, M Schafers, M Marziniak, K.V Toyka, Etanercept reduces hyperalgesia in experimental painful neuropathy, J Peripher Nerv Syst (2001) 67-72 C Sommer, M Kress, Recent findings on how proinflammatory cytokines cause pain: peripheral mechanisms in inflammatory and neuropathic hyperalgesia, Neurosci Lett 361 (2004) 184-187 L.S Sorkin, M Schafers, Immune Cells in Peripheral Nerve, IASP Press, Seattle (2007) S.J Stankus, M Dlugopolski, D Packer, Management of herpes zoster (shingles) and postherpetic neuralgia, Am Fam Physician 61:8 (2000) 2437-2444 G.H Stein, T98G: an anchorage-independent human tumor cell line that exhibits stationary phase G1 arrest in vitro, J Cell Physiol 99 (1979) 43-54 J.M Walker, P.J Sweeney, Production of protein hydrolysates using enzymes, In: The Protein Protocols Handbook, 2nd Edition, Ed: J.M Walker, Humana Press Inc, Totowa (2002) J.P Syriatowicz, D Hu, J.S Walker, D.J Tracey, Hyperalgesia due to nerve injury: role of prostaglandins, Neuroscience 94:2 (1999) 587-594 S Tachibana, R Sethuraman, T.L Lee, Relationships between physiological amino acids in cerebrospinal fluid and pain, Recent Dev Pain Res (2005) 95-114 F.Y Tanga, V Raghavendra, J.A DeLeo, Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain, Neurochem Intl 45 (2004) 397-407 116 R Vallejo, D.M Tilly, L Vogel, R Benyamin, The role of glia and the immune system in the development and maintenance of neuropathic pain, Pain Prac 10:3 (2010) 167-184 N.J Van Wagoner, J.-W Oh, P Repovic, E.N Benveniste, Interleukin-6 (IL-6) production by astrocytes: autocrine regulation by IL-6 and the soluble IL-6 receptor, J Neurosci 19:13 (1999) 5236–5244 A.J van Wijick, W Opstelten, K.G Moon, G.A van Essen, R.J Stolker, C.J Kalkman, T.J Verheij, The PINE study of epidural steroids and local anesthetics to prevent postherpetic neuralgia: a randomized controlled trial, Lancet 367 (2006) 219- 224 A Verkhratsky, C Steinhauser, Ion channels in glial cells, Brain Res Rev 32 (2000) 380-412 R Wagner, R.R Myers, Endoneurial injection of TNF-α produces neuropathic pain behaviors, Neuroreport (1996) 2897-2901 L.R Watkins, S.F Maier, Beyond neurons: evidence that immune and glial cells contribute to pathological pain states, Physiol Rev 82 (2002) 981-1011 R.J Whitley, H.L Weiss, S.J Soong, J.W Gnann, Herpes zoster: risk categories for persistent pain, J Infect Dis 179 (1999) 9-15 F Wiederkehr, Analysis of cerebrospinal fluid proteins by electrophoresis, J Chromatogr 569 (1991) 281-296 J Wieseler-Frank, S.F Maier, L.R Watkins, Glial activation and pathological pain, Neurochem Intl 45 (2004) 389-395 J Wieseler-Frank, S.F Maier, L.R Watkins, Central proinflammatory cytokines and pain enhancement, Neurosignals 14 (2005) 166-174 C.L Wu, A Marsh, R.H Dworkin, The role of sympathetic nerve blocks in herpes zoster and postherpetic neuralgia, Pain 87 (2000) 121-129 T Yamamoto, T.L Yaksh, Effects of intrathecal strychnine and bicuculline on nerve compression-induced thermal hyperalgesia and selective antagonism by MK-801, Pain 54 (1993) 79–84 S.N Yeo, K.H Tay, Pain prevalence in Singapore, Ann Acad Med Singapore 38:11 (2009) 937-942 T.M Zanjani, M Sabetkasaei, N Mosaffa, H Manaheji, F Labibi, B Farokhi, Suppression of interleukin-6 by minocycline in a rat model of neuropathic pain, Eur J Pharmacol 538 (2006) 66-72 117 J Zhang, D.R Goodlett, T.J Montine, Proteomic biomarker discovery in cerebrospinal fluid for neurodegenerative diseases, J Alzheimers Dis (2005) 377–386 A Zougman, B Pilch, A Podtelejnikov,M Kiehntopf, C Schnabel, C Kumar, M Mann, Integrated analysis of the cerebrospinal fluid peptidome and proteome, J Proteome Res (2008) 386–399 J.W Zumwalt, B.J Thunstrom, B.L Spangelo, Interleukin-1β and catecholamines synergistically stimulate interleukin-6 release from rat C6 glioma Cells in vitro: a potential role for lysophosphatidylcholine, Endocrinol 140:2 (1999) 888-896 118 ... aspartate, arginine, asparagine, tyrosine and valine were also demonstrated to be elevated in the CSF of chronic pain patients as compared to that of acute pain patients On the other hand, GABA... microglia are involved in the early development of chronic pain, while astrocytes function in sustaining the pain (Vallejo et al., 2010) An interesting finding was that nerve injury induces an increase... Pain The International Association for the Study of Pain defines chronic pain as pain that persists for at least months Chronic pain is normally triggered by injury or disease, which damages the

Ngày đăng: 02/10/2015, 17:15

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan