リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Development of Remote Inflammation through Interneuron Network in the Spinal Cord」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Development of Remote Inflammation through Interneuron Network in the Spinal Cord

HALAKA, Nada Nasr Abdelmoneim Sayed Ahmed 北海道大学

2022.03.24

概要

Background and purpose: Rheumatoid arthritis (RA) is a chronic inflammatory disease accompanied by joint inflammation, synovial hypertrophy, and progressive destruction of cartilage and bone, and affects 1% of the adult population worldwide. One criterion of the diagnosis and a fundamental characteristic of RA is remote inflammation, which results in widespread and severe malformation and immobility on both sides of joint. Although several studies have suggested that a neural mechanism is involved in the spreading inflammation of RA, the molecular links of inflammatory and neural pathways have not been uncovered. We previously found an amplification mechanism of inflammatory response, so called "IL-6 amplifier", in which a simultaneous activation of IL-6-STAT3 and other cytokines (e.g. IL-17, TNF-α, and growth factors)-NF-κB pathways, induces an excessive activation of NF-κB in tissue-specific non- immune cells, resulting in chronic inflammation through a local recruitment of immune cells and robust production of inflammatory cytokines such as IL-6 per se and chemokines. Accordingly, we have defined inflammation as an IL-6-mediated accumulation of immune cells, and/or proliferation of immune cells and regional non-immune cells, followed by the dysregulation of local homeostasis including the dysfunction of tissues and/or organs. Furthermore, we also discovered “gateway reflex” as the regulatory mechanism of IL-6 amplifier by activation of specific neural circuit. Environmental factors such as gravity, weak electricity, pain, stress, and light stimulate specific neural circuits to produce neurotransmitter locally, and induce or inhibit (only by light stimulation) IL-6 amplifier that subsequently creates a specific gateway for immune cells at blood barrier, and then promote or suppress (only by light stimulation) an onset of chronic inflammatory diseases. Based on our original discoveries regarding IL-6 amplifier and gateway reflex, we hypothesized that a local neural regulation for bilateral inflammation may trigger initial inflammation and promotes the subsequent recruitment of immune cells by releasing attracting factors such as chemokines via the IL-6 amplifier. In my doctoral research, I aimed to clarify the underlying molecular mechanism of how specific neural circuit interconnects with bilateral inflammation for spreading pathology.

Materials and methods: To explore the molecular basis via neural pathways for spreading inflammation, F759 mice in which IL-6-mediated STAT3 activation is enhanced due to an amino acid substitution, Y759F in gp130 (IL-6 signal transducer in the IL-6R complex) at the binding site of SOCS3 for the negative regulation of IL-6 signaling, and spontaneous development of an RA-like disease in both sides of the ankle joint at around one year of age occur, were used as a representative cytokine-induced arthritis model as well as in a collagen-induced arthritis model (CIA) throughout my study.
Since we hypothesized that spreading inflammation in RA might depend on neural pathways distributed on both sides, F759 mice were injected with IL-6 plus IL-17A, saline, or a neurotransmitter, adenosine triphosphate (ATP), with or without anti-IL-6R, anti-IL-17A, control IgG, A438079 (ATP receptor P2X7 antagonist) at unilateral or bilateral ankle joints. For some experiments, F759 mice induced arthritis by cytokine injections were deafferented the sensory neurons at one side of the dorsal root ganglions (DRGs) beside the fifth lumbar cord (L5) or spinal cord cut. These mice were then assessed for the disease severity based on two bilaterally parameters: (1) swelling of the ankle and (2) restricted mobility of the ankle joints. The severity of each parameter was graded on a scale of 0–3, where 0 indicates no change; 1, mild change; 2, medium change; and 3, severe change. Averages for a single point in one leg ankle joint from each mouse were used. Ipsilateral and/or contralateral ankle joints, DRG at third to sixth lumbar cords (L3-L6) and nineth to thirteenth thoracic cords (T9-13), were used for examining recruitment of immune cells and neural activation status, and identification of specific sensory neuron and interneuron by immunohistochemistry (IHC), for quantifying c-fos (neural activation status), cytokine, and chemokine expression levels by real-time PCR, and for measuring cytokine and neurotransmitter levels by ELISA. For assessing NF-κB activity, ankle joints from NF-κB-reporter Tg/F759 mice induced arthritis by IL-6 plus IL-17A injections were collected, and synovial tissues were homogenized in passive lysis buffer. After centrifugation, the supernatants were collected and analyzed for luciferase activities using luciferase reporter assay system. To investigate the neural networks in spinal cord that connect sensory pathways between the ankle joints, herpes simplex virus 2 (HSV2) was employed to trace neural connections regardless of the presence of synapses. The presence of HSV2 was detected by IHC and quantified by real-time PCR.

Results: The surgical ablation or pharmacological inhibition of neural pathway at one side of ankle prevented inflammation development on the other side. Mechanistic analysis showed that ATP induced by activation of the IL-6 amplifier in collagen type 1+ non-immune cells in one side of the ankle joint activates Nav1.8+ TRPV1+/- sensory neurons, which further stimulate the regional sensory neural pathway involving the lower thoracic spinal cord that contains proenkephalin+ interneurons. On the contralateral side, the response of sensory neurons in L4-L6 DRGs releases ATP, which induces inflammatory mediators, including cytokines and chemokines, by activating the IL-6 amplifier in collagen type 1+ non-immune cells, including fibroblasts and endothelial cells, through a ATP receptor, P2RX7.

Discussion: It is known that spreading inflammation is common in RA. The present study clarifies the molecular link for spreading inflammation between bilateral ankle joints in cytokine-induced and CIA models. Cytokine injections into one ankle joint of F759 mice induce ATP release from non-immune cells and subsequently activate afferent sensory pathways with c-fos induction on both sides of L5 DRG toward the contralateral ankle joint through a proenkephalin+ interneuron network in the thoracic cords, suggesting that the inflammation signal from one side activates sensory neurons in both the ipsilateral and contralateral sides. A previous report showed that RA synovial fibroblasts (RASFs) are able to migrate and contribute to the spread of the disease between bilateral synovial tissues in SCID mice. Although the authors identified RASFs as one key factor for spreading inflammation from a single joint to multiple ones, how RASFs reach the other joints remains to be elucidated. The present study suggests that the local neural regulation for bilateral inflammation may trigger the initial inflammation and promotes the subsequent recruitment of RASFs by releasing chemokines via IL-6 amplifier. Thus, a regional sensory neuron-interneuron connection between the ankle joints through the thoracic spinal cord is critical for spreading inflammation via the bilateral expression of ATP, which activates a neural pathway and enhances the IL-6 amplifier.

Conclusion: My doctoral study suggests that blockade of the sensory neuron-interneuron axis may be a therapeutic target for various inflammatory diseases including RA, in which inflammation spreads to remote positions.

参考文献

Arima, Y., Harada, M., Kamimura, D., Park, J.H., Kawano, F., Yull, F.E., Kawamoto, T., Iwakura, Y., Betz, U.A., Márquez, G., et al. (2012). Regional neural activation defines a gateway for autoreactive T cells to cross the blood-brain barrier. Cell 148, 447-457. 10.1016/j.cell.2012.01.022.

Arima, Y., Kamimura, D., Atsumi, T., Harada, M., Kawamoto, T., Nishikawa, N., Stofkova, A., Ohki, T., Higuchi, K., Morimoto, Y., et al. (2015a). A pain-mediated neural signal induces relapse in murine autoimmune encephalomyelitis, a multiple sclerosis model. eLife 4, e08733. 10.7554/eLife.08733.

Arima, Y., Kamimura, D., Atsumi, T., Harada, M., Kawamoto, T., Nishikawa, N., Stofkova, A., Ohki, T., Higuchi, K., Morimoto, Y., et al. (2015b). A pain-mediated neural signal induces relapse in murine autoimmune encephalomyelitis, a multiple sclerosis model. Elife 4. 10.7554/eLife.08733.

Arima, Y., Ohki, T., Nishikawa, N., Higuchi, K., Ota, M., Tanaka, Y., Nio-Kobayashi, J., Elfeky, M., Sakai, R., Mori, Y., et al. (2017a). Brain micro-inflammation at specific vessels dysregulates organ-homeostasis via the activation of a new neural circuit. eLife 6, e25517. 10.7554/eLife.25517.

Arima, Y., Ohki, T., Nishikawa, N., Higuchi, K., Ota, M., Tanaka, Y., Nio-Kobayashi, J., Elfeky, M., Sakai, R., Mori, Y., et al. (2017b). Brain micro-inflammation at specific vessels dysregulates organ-homeostasis via the activation of a new neural circuit. Elife 6. 10.7554/eLife.25517.

Arnett, F.C., Edworthy, S.M., Bloch, D.A., McShane, D.J., Fries, J.F., Cooper, N.S., Healey, L.A., Kaplan, S.R., Liang, M.H., and Luthra, H.S. (1988). The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 31, 315-324.

Arulkumaran, N., Unwin, R.J., and Tam, F.W. (2011). A potential therapeutic role for P2X7 receptor (P2X7R) antagonists in the treatment of inflammatory diseases. Expert Opin Investig Drugs 20, 897-915. 10.1517/13543784.2011.578068.

Atsumi, T., Ishihara, K., Kamimura, D., Ikushima, H., Ohtani, T., Hirota, S., Kobayashi, H., Park, S.J., Saeki, Y., Kitamura, Y., and Hirano, T. (2002). A point mutation of Tyr-759 in interleukin 6 family cytokine receptor subunit gp130 causes autoimmune arthritis. J Exp Med 196, 979-990. 10.1084/jem.20020619.

Atsumi, T., Singh, R., Sabharwal, L., Bando, H., Meng, J., Arima, Y., Yamada, M., Harada, M., Jiang, J.J., Kamimura, D., et al. (2014). Inflammation amplifier, a new paradigm in cancer biology. Cancer Res 74, 8-14. 10.1158/0008-5472.Can-13-2322.

Atsumi, T., Suzuki, H., Jiang, J.J., Okuyama, Y., Nakagawa, I., Ota, M., Tanaka, Y., Ohki, T., Katsunuma, K., Nakajima, K., et al. (2017). Rbm10 regulates inflammation development via alternative splicing of Dnmt3b. Int Immunol 29, 581-591. 10.1093/intimm/dxx067.

Biggioggero, M., Crotti, C., Becciolini, A., and Favalli, E.G. (2019). Tocilizumab in the treatment of rheumatoid arthritis: an evidence-based review and patient selection. Drug Des Devel Ther 13, 57-70. 10.2147/dddt.S150580.

Bliss, T.V., Collingridge, G.L., Kaang, B.K., and Zhuo, M. (2016). Synaptic plasticity in the anterior cingulate cortex in acute and chronic pain. Nat Rev Neurosci 17, 485-496. 10.1038/nrn.2016.68.

Burnstock, G. (2006). Historical review: ATP as a neurotransmitter. Trends Pharmacol Sci 27, 166-176. 10.1016/j.tips.2006.01.005.

Chavan, S.S., Pavlov, V.A., and Tracey, K.J. (2017). Mechanisms and Therapeutic Relevance of Neuro-immune Communication. Immunity 46, 927-942. 10.1016/j.immuni.2017.06.008.

Clarke, G.S., Buckland-Wright, J.C., and Grahame, R. (1994 ). Symmetry of radiological features in the wrist and hands of patients with early to moderate rheumatoid arthritis: a quantitative microfocal radiographic study. Br J Rheumatol. 33, 249-254.

da Silva, J.L.G., Passos, D.F., Bernardes, V.M., and Leal, D.B.R. (2019). ATP and adenosine: Role in the immunopathogenesis of rheumatoid arthritis. Immunol Lett 214, 55-64. 10.1016/j.imlet.2019.08.009.

Donaldson, L.F., McQueen, D.S., and Seckl, J.R. (1995). Neuropeptide gene expression and capsaicin-sensitive primary afferents: maintenance and spread of adjuvant arthritis in the rat. J Physiol 486 ( Pt 2), 473-482.

Donnelly-Roberts, D.L., Namovic, M.T., Han, P., and Jarvis, M.F. (2009). Mammalian P2X7 receptor pharmacology: comparison of recombinant mouse, rat and human P2X7 receptors. Br J Pharmacol. 157, 1203-1214.

Evans, R.J., Derkach, V., and Surprenant, A. (1992). ATP mediates fast synaptic transmission in mammalian neurons. Nature 357, 503-505.

Fan, Z.D., Zhang, Y.Y., Guo, Y.H., Huang, N., Ma, H.H., Huang, H., and Yu, H.G. (2016). Involvement of P2X7 receptor signaling on regulating the differentiation of Th17 cells and type II collagen-induced arthritis in mice. Sci Rep 6, 35804. 10.1038/srep35804.

Feldmann, M., Brennan, F.M., and Maini, R.N. (1996). Rheumatoid arthritis. Cell 85, 307-310. 10.1016/s0092-8674(00)81109-5.

Finger, T.E., Danilova, V., Barrows, J., Bartel, D.L., Vigers, A.J., Stone, L., Hellekant, G., and Kinnamon, S.C. (2005). ATP signaling is crucial for communication from taste buds to gustatory nerves. Science 10, 1495-1499.

Fujita, M., Yamamoto, Y., Jiang, J.J., Atsumi, T., Tanaka, Y., Ohki, T., Murao, N., Funayama, E., Hayashi, T., Osawa, M., et al. (2019). NEDD4 Is Involved in Inflammation Development during Keloid Formation. J Invest Dermatol 139, 333-341. 10.1016/j.jid.2018.07.044.

Fukada, T., Hibi, M., Yamanaka, Y., Takahashi-Tezuka, M., Fujitani, Y., Yamaguchi, T., Nakajima, K., and Hirano, T. (1996). Two signals are necessary for cell proliferation induced by a cytokine receptor gp130: involvement of STAT3 in anti-apoptosis. Immunity 5, 449-460. 10.1016/s1074-7613(00)80501-4.

Gogan, P., Gueritaud, J.P., and Tyc-Dumont, S. (1983). Comparison of antidromic and orthodromic action potentials of identified motor axons in the cat's brain stem. J Physiol 335, 205-220.

Gourine, A.V., Llaudet, E., Dale, N., and Spyer, K.M. (2005). ATP is a mediator of chemosensory transduction in the central nervous system. Nature. 436, 108-111.

Grassel, S.G. (2014). The role of peripheral nerve fibers and their neurotransmitters in cartilage and bone physiology and pathophysiology. Arthritis Res Ther 16, 485.

Harada, M., Kamimura, D., Arima, Y., Kohsaka, H., Nakatsuji, Y., Nishida, M., Atsumi, T., Meng, J., Bando, H., Singh, R., et al. (2015). Temporal expression of growth factors triggered by epiregulin regulates inflammation development. Journal of immunology 194, 1039-1046. 10.4049/jimmunol.1400562.

Hashizume, M., and Mihara, M. (2011). The roles of interleukin-6 in the pathogenesis of rheumatoid arthritis. Arthritis 2011, 765624. 10.1155/2011/765624.

Higuchi, H., Kamimura, D., Jiang, J.J., Atsumi, T., Iwami, D., Hotta, K., Harada, H., Takada, Y., Kanno-Okada, H., Hatanaka, K.C., et al. (2020). Orosomucoid 1 is involved in the development of chronic allograft rejection after kidney transplantation. Int Immunol 32, 335-346. 10.1093/intimm/dxaa003.

Hirano, T. (1992). Interleukin-6 and its relation to inflammation and disease. Clin Immunol Immunopathol 62, S60-65. 10.1016/0090-1229(92)90042-m.

Hirano, T. (1998). Interleukin 6 and its receptor: ten years later. Int Rev Immunol 16, 249-284. 10.3109/08830189809042997.

Hirano, T., Matsuda, T., Turner, M., Miyasaka, N., Buchan, G., Tang, B., Sato, K., Shimizu, M., Maini, R., Feldmann, M., and et al. (1988). Excessive production of interleukin 6/B cell stimulatory factor-2 in rheumatoid arthritis. Eur J Immunol 18, 1797-1801. 10.1002/eji.1830181122.

Hirano, T., and Murakami, M. (2020). COVID-19: A New Virus, but a Familiar Receptor and Cytokine Release Syndrome. Immunity 52, 731-733. 10.1016/j.immuni.2020.04.003.

Hirano, T., Taga, T., Yasukawa, K., Nakajima, K., Nakano, N., Takatsuki, F., Shimizu, M., Murashima, A., Tsunasawa, S., Sakiyama, F., and et al. (1987). Human B-cell differentiation factor defined by an anti-peptide antibody and its possible role in autoantibody production. Proceedings of the National Academy of Sciences of the United States of America 84, 228-231. 10.1073/pnas.84.1.228.

Holton, P. (1959). The liberation of adenosine triphosphate on antidromic stimulation of sensory nerves. J Physiol 145, 494-504.

Houssiau, F.A., Devogelaer, J.P., Van Damme, J., de Deuxchaisnes, C.N., and Van Snick, J. (1988). Interleukin-6 in synovial fluid and serum of patients with rheumatoid arthritis and other inflammatory arthritides. Arthritis Rheum 31, 784-788. 10.1002/art.1780310614.

Igarashi, H., Hashimoto, J., Tomita, T., Yoshikawa, H., and Ishihara, K. (2010). TP53 mutations coincide with the ectopic expression of activation-induced cytidine deaminase in the fibroblast-like synoviocytes derived from a fraction of patients with rheumatoid arthritis. Clin Exp Immunol 161, 71-80. 10.1111/j.1365-2249.2010.04163.x.

Ishihara, K., and Hirano, T. (2002). IL-6 in autoimmune disease and chronic inflammatory proliferative disease. Cytokine Growth Factor Rev 13, 357-368. 10.1016/s1359- 6101(02)00027-8.

Jørgensen, N.R. (2018). The purinergic P2X7 ion channel receptor-a 'repair' receptor in bone. Curr Opin Immunol 52, 32-38. 10.1016/j.coi.2018.03.016.

June, R.R., and Olsen, N.J. (2016). Room for more IL-6 blockade? Sarilumab for the treatment of rheumatoid arthritis. Expert Opin Biol Ther 16, 1303-1309. 10.1080/14712598.2016.1217988.

Kamimura, D., Ishihara, K., and Hirano, T. (2003). IL-6 signal transduction and its physiological roles: the signal orchestration model. Rev Physiol Biochem Pharmacol 149, 1-38. 10.1007/s10254-003-0012-2.

Kawamoto, T., and Kawamoto, K. (2014). Preparation of thin frozen sections from nonfixed and undecalcified hard tissues using Kawamot's film method (2012). Methods Mol Biol 1130, 149-164. 10.1007/978-1-62703-989-5_11.

Kawano, F., Ishihara, A., Stevens, J.L., Wang, X.D., Ohshima, S., Horisaka, M., Maeda, Y., Nonaka, I., and Ohira, Y. (2004). Tension- and afferent input-associated responses of neuromuscular system of rats to hindlimb unloading and/or tenotomy. Am J Physiol Regul Integr Comp Physiol 287, R76-86. 10.1152/ajpregu.00694.2003.

Kawano, F., Matsuoka, Y., Oke, Y., Higo, Y., Terada, M., Wang, X.D., Nakai, N., Fukuda, H., Imajoh-Ohmi, S., and Ohira, Y. (2007). Role(s) of nucleoli and phosphorylation of ribosomal protein S6 and/or HSP27 in the regulation of muscle mass. American journal of physiology. Cell physiology 293, C35-44. 10.1152/ajpcell.00297.2006.

Kelly, S., Dunham, J.P., and Donaldson, L.F. (2007). Sensory nerves have altered function contralateral to a monoarthritis and may contribute to the symmetrical spread of inflammation. Eur J Neurosci 26, 935-942. 10.1111/j.1460-9568.2007.05737.x.

Kidd, B.L., Mapp, P.I., Gibson, S.J., Polak, J.M., O'Higgins, F., Buckland-Wright, J.C., and Blake, D.R. (1989). A neurogenic mechanism for symmetrical arthritis. Lancet 2, 1128-1130.

Kim, S.S., Wang, H., Li, X.Y., Chen, T., Mercaldo, V., Descalzi, G., Wu, L.J., and Zhuo, M. (2011). Neurabin in the anterior cingulate cortex regulates anxiety-like behavior in adult mice. Mol Brain. 19, 4-6.

Kitabayashi, C., Fukada, T., Kanamoto, M., Ohashi, W., Hojyo, S., Atsumi, T., Ueda, N., Azuma, I., Hirota, H., Murakami, M., and Hirano, T. (2010). Zinc suppresses Th17 development via inhibition of STAT3 activation. . Int. Immunol. 22, 375-386.

Labasi, J.M., Petrushova, N., Donovan, C., McCurdy, S., Lira, P., Payette, M.M., Brissette, W., Wicks, J.R., Audoly, L., and Gabel, C.A. (2002). Absence of the P2X7 receptor alters leukocyte function and attenuates an inflammatory response. Journal of immunology 168, 6436-6445. 10.4049/jimmunol.168.12.6436.

Larsson, J., Ekblom, A., Henriksson, K., Lundeberg, T., and Theodorsson, E. (1991). Concentration of substance P, neurokinin A, calcitonin gene-related peptide, neuropeptide Y and vasoactive intestinal polypeptide in synovial fluid from knee joints in patients suffering from rheumatoid arthritis. Scand J Rheumatol 20, 326-335.

Lee, J., Nakagiri, T., Kamimura, D., Harada, M., Oto, T., Susaki, Y., Shintani, Y., Inoue, M., Miyoshi, S., Morii, E., et al. (2013). IL-6 amplifier activation in epithelial regions of bronchi after allogeneic lung transplantation. Int Immunol 25, 319-332. 10.1093/intimm/dxs158.

Lefevre, S., Knedla, A., Tennie, C., Kampmann, A., Wunrau, C., Dinser, R., Korb, A., Schnaker, E.M., Tarner, I.H., Robbins, P.D., et al. (2009). Synovial fibroblasts spread rheumatoid arthritis to unaffected joints. Nat Med 15, 1414-1420. 10.1038/nm.2050.

Lopez-Castejon, G., Theaker, J., Pelegrin, P., Clifton, A.D., Braddock, M., and Surprenant, A. (2010). P2X(7) receptor-mediated release of cathepsins from macrophages is a cytokine- independent mechanism potentially involved in joint diseases. J Immunol 185, 2611-2619. 10.4049/jimmunol.1000436.

Malmberg, A.B., and Basbaum, A.I. (1998). Partial sciatic nerve injury in the mouse as a model of neuropathic pain: behavioral and neuroanatomical correlates. Pain 76, 215-222.

Mausset-Bonnefont, A.L., Cren, M., Vicente, R., Quentin, J., Jorgensen, C., Apparailly, F., and Louis-Plence, P. (2019). Arthritis sensory and motor scale: predicting functional deficits from the clinical score in collagen-induced arthritis. Arthritis Res Ther 21, 264. 10.1186/s13075-019-2047-z.

McGaraughty, S., Chu, K.L., Namovic, M.T., Donnelly-Roberts, D.L., Harris, R.R., Zhang, X.F., Shieh, C.C., Wismer, C.T., Zhu, C.Z., Gauvin, D.M., et al. (2007). P2X7-related modulation of pathological nociception in rats. Neuroscience. 148, 1817-1828.

Meng, J., Jiang, J.J., Atsumi, T., Bando, H., Okuyama, Y., Sabharwal, L., Nakagawa, I., Higuchi, H., Ota, M., Okawara, M., et al. (2016). Breakpoint Cluster Region-Mediated Inflammation Is Dependent on Casein Kinase II. Journal of immunology 197, 3111-3119. 10.4049/jimmunol.1601082.

Miller, L.E., Grifka, J., Scholmerich, J., and Straub, R.H. (2002). Norepinephrine from synovial tyrosine hydroxylase positive cells is a strong indicator of synovial inflammation in rheumatoid arthritis. J Rheumatol 29, 427-435.

Moretto, J.N., Duffy Á, M., and Scharfman, H.E. (2017). Acute restraint stress decreases c- fos immunoreactivity in hilar mossy cells of the adult dentate gyrus. Brain Struct Funct 222, 2405-2419. 10.1007/s00429-016-1349-z.

Murakami, K., Tanaka, Y., and Murakami, M. (2021). The gateway reflex: breaking through the blood barriers. Int Immunol. 10.1093/intimm/dxab064.

Murakami, M., Harada, M., Kamimura, D., Ogura, H., Okuyama, Y., Kumai, N., Okuyama, A., Singh, R., Jiang, J.J., Atsumi, T., et al. (2013). Disease-association analysis of an inflammation-related feedback loop. Cell Rep 3, 946-959. 10.1016/j.celrep.2013.01.028.

Murakami, M., Kamimura, D., and Hirano, T. (2019). Pleiotropy and Specificity: Insights from the Interleukin 6 Family of Cytokines. Immunity 50, 812-831. 10.1016/j.immuni.2019.03.027.

Murakami, M., Okuyama, Y., Ogura, H., Asano, S., Arima, Y., Tsuruoka, M., Harada, M., Kanamoto, M., Sawa, Y., Iwakura, Y., et al. (2011). Local microbleeding facilitates IL-6- and IL-17-dependent arthritis in the absence of tissue antigen recognition by activated T cells. J Exp Med 208, 103-114. jem.20100900 [pii]10.1084/jem.20100900.

Nakagawa, T., Tsuruoka, M., Ogura, H., Okuyama, Y., Arima, Y., Hirano, T., and Murakami, M. (2010). IL-6 positively regulates Foxp3+CD8+ T cells in vivo. Int Immunol 22, 129-139. 10.1093/intimm/dxp119.

Narváez, J., Oton, T., J, L.L., Mora-Limiñana, M., Nolla, J.M., and Loza, E. (2021). Response to interleukin-6 receptor antagonists in patients with rheumatoid arthritis is independent of the number of prior used TNF inhibitors: A systematic review and metaanalysis. Joint Bone Spine 88, 105112. 10.1016/j.jbspin.2020.105112.

Niu, Q., Cai, B., Huang, Z.C., Shi, Y.Y., and Wang, L.L. (2012). Disturbed Th17/Treg balance in patients with rheumatoid arthritis. Rheumatol Int 32, 2731-2736. 10.1007/s00296- 011-1984-x.

Norgren, R.B.J., and Lehman, M.N. (1998). Herpes simplex virus as a transneuronal tracer. Neurosci Biobehav Rev. 22, 695-708.

O'Connor, T.M., O'Connell, J., O'Brien, D.I., Goode, T., Bredin, C.P., and Shanahan, F. (2004). The role of substance P in inflammatory disease. J Cell Physiol 201, 167-180. 10.1002/jcp.20061.

Ogata, A., Kato, Y., Higa, S., and Yoshizaki, K. (2019). IL-6 inhibitor for the treatment of rheumatoid arthritis: A comprehensive review. Mod Rheumatol 29, 258-267. 10.1080/14397595.2018.1546357.

Ogura, H., Murakami, M., Okuyama, Y., Tsuruoka, M., Kitabayashi, C., Kanamoto, M., Nishihara, M., Iwakura, Y., and Hirano, T. (2008). Interleukin-17 promotes autoimmunity by triggering a positive-feedback loop via interleukin-6 induction. Immunity 29, 628-636. 10.1016/j.immuni.2008.07.018.

Ohtani, T., Ishihara, K., Atsumi, T., Nishida, K., Kaneko, Y., Miyata, T., Itoh, S., Narimatsu, M., Maeda, H., Fukada, T., et al. (2000). Dissection of signaling cascades through gp130 in vivo: reciprocal roles for STAT3- and SHP2-mediated signals in immune responses. Immunity 12, 95-105. 10.1016/s1074-7613(00)80162-4.

Okuyama, Y., Tanaka, Y., Jiang, J.J., Kamimura, D., Nakamura, A., Ota, M., Ohki, T., Higo, D., Ogura, H., Ishii, N., et al. (2018a). Bmi1 Regulates IkappaBalpha Degradation via Association with the SCF Complex. Journal of immunology 201, 2264-2272. 10.4049/jimmunol.1701223.

Okuyama, Y., Tanaka, Y., Jiang, J.J., Kamimura, D., Nakamura, A., Ota, M., Ohki, T., Higo, D., Ogura, H., Ishii, N., et al. (2018b). Bmi1 Regulates IκBα Degradation via Association with the SCF Complex. J Immunol 201, 2264-2272. 10.4049/jimmunol.1701223.

Ota, M., Tanaka, Y., Nakagawa, I., Jiang, J.J., Arima, Y., Kamimura, D., Onodera, T., Iwasaki, N., and Murakami, M. (2020). Role of Chondrocytes in the Development of Rheumatoid Arthritis Via Transmembrane Protein 147-Mediated NF-kappaB Activation. Arthritis Rheumatol 72, 931-942. 10.1002/art.41182.

Peirs, C., Williams, S.P., Zhao, X., Walsh, C.E., Gedeon, J.Y., Cagle, N.E., Goldring, A.C., Hioki, H., Liu, Z., Marell, P.S., and Seal, R.P. (2015). Dorsal Horn Circuits for Persistent Mechanical Pain. Neuron 87, 797-812. 10.1016/j.neuron.2015.07.029.

Raimondo, M.G., Biggioggero, M., Crotti, C., Becciolini, A., and Favalli, E.G. (2017). Profile of sarilumab and its potential in the treatment of rheumatoid arthritis. Drug Des Devel Ther 11, 1593-1603. 10.2147/dddt.S100302.

Russell, F.A., Schuelert, N., Veldhoen, V.E., Hollenberg, M.D., and McDougall, J.J. (2012). Activation of PAR(2) receptors sensitizes primary afferents and causes leukocyte rolling and adherence in the rat knee joint. Br J Pharmacol 167, 1665-1678. 10.1111/j.1476- 5381.2012.02120.x.

Sadikot, R.T., and Blackwell, T.S. (2008). Bioluminescence: imaging modality for in vitro and in vivo gene expression. Methods Mol Biol 477, 383-394. 10.1007/978-1-60327-517- 0_29.

Sasai, M., Saeki, Y., Ohshima, S., Nishioka, K., Mima, T., Tanaka, T., Katada, Y., Yoshizaki, K., Suemura, M., and Kishimoto, T. (1999). Delayed onset and reduced severity of collagen-induced arthritis in interleukin-6-deficient mice. Arthritis Rheum. 42, 1635-1643. Sawa, S., Kamimura, D., Jin, G.H., Morikawa, H., Kamon, H., Nishihara, M., Ishihara, K., Murakami, M., and Hirano, T. (2006). Autoimmune arthritis associated with mutated interleukin (IL)-6 receptor gp130 is driven by STAT3/IL-7-dependent homeostatic proliferation of CD4+ T cells. J Exp Med 203, 1459-1470. 10.1084/jem.20052187.

Schmidt-Supprian, M., Bloch, W., Courtois, G., Addicks, K., Israël, A., Rajewsky, K., and Pasparakis, M. (2000). NEMO/IKK gamma-deficient mice model incontinentia pigmenti. Mol . Cell 5, 981-992.

Seltzer, Z., Dubner, R., and Shir, Y. (1990). A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 43, 205-218.

Shenker, N., Haigh, R., Roberts, E., Mapp, P., Harris, N., and Blake, D. (2003). A review of contralateral responses to a unilateral inflammatory lesion. Rheumatology (Oxford). 42, 1279-1286.

Sorkin, L.S., Eddinger, K.A., Woller, S.A., and Yaksh, T.L. (2018). Origins of antidromic activity in sensory afferent fibers and neurogenic inflammation. Semin Immunopathol 40, 237-247. 10.1007/s00281-017-0669-2.

Stofkova, A., Kamimura, D., Ohki, T., Ota, M., Arima, Y., and Murakami, M. (2019). Photopic light-mediated down-regulation of local α(1A)-adrenergic signaling protects blood- retina barrier in experimental autoimmune uveoretinitis. Sci Rep 9, 2353. 10.1038/s41598- 019-38895-y.

Stotz-Potter, E.H., Morin, S.M., and DiMicco, J.A. (1996). Effect of microinjection of muscimol into the dorsomedial or paraventricular hypothalamic nucleus on air stress-induced neuroendocrine and cardiovascular changes in rats. Brain Res 742, 219-224. 10.1016/s0006- 8993(96)01011-6.

Takeda, K., Kaisho, T., Yoshida, N., Takeda, J., Kishimoto, T., and Akira, S. (1998). Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: generation and characterization of T cell-specific Stat3-deficient mice. J Immunol 161, 4652- 4660.

Talbot, S., Foster, S.L., and Woolf, C.J. (2016). Neuroimmunity: Physiology and Pathology. Annu Rev Immunol 34, 421-447. 10.1146/annurev-immunol-041015-055340.

Tanaka, Y., Arima, Y., Higuchi, K., Ohki, T., Elfeky, M., Ota, M., Kamimura, D., and Murakami, M. (2017). EAE Induction by Passive Transfer of MOG-specific CD4(+) T Cells. Bio Protoc 7, e2370. 10.21769/BioProtoc.2370.

Tanaka, Y., Sabharwal, L., Ota, M., Nakagawa, I., Jiang, J.J., Arima, Y., Ogura, H., Okochi, M., Ishii, M., Kamimura, D., and Murakami, M. (2018). Presenilin 1 Regulates NF-kappaB Activation via Association with Breakpoint Cluster Region and Casein Kinase II. Journal of immunology 201, 2256-2263. 10.4049/jimmunol.1701446.

Turner, S.L., and Jenkins, F.J. (1997). The roles of herpes simplex virus in neuroscience. J Neurovirol. 3, 110-125.

Uchida, M., Yamamoto, R., Matsuyama, S., Murakami, K., Hasebe, R., Hojyo, S., Tanaka, Y., and Murakami, M. (2021). Gateway reflexes, neuronal circuits that regulate the gateways for autoreactive T cells in organs that have blood barriers. Int Immunol. 10.1093/intimm/dxab022.

Vetrugno, G.C., Lachuer, J., Perego, C., Miranda, E., De Simoni, M.G., and Tappaz, M. (1993). Lack of glucocorticoids sustains the stress-induced release of noradrenaline in the anterior hypothalamus. Neuroendocrinology 57, 835-842. 10.1159/000126442.

Xie, Z., Dai, J., Yang, A., and Wu, Y. (2014). A role for bradykinin in the development of anti-collagen antibody-induced arthritis. Rheumatology (Oxford) 53, 1301-1306. 10.1093/rheumatology/keu015.

Yoshizaki, K., Matsuda, T., Nishimoto, N., Kuritani, T., Taeho, L., Aozasa, K., Nakahata, T., Kawai, H., Tagoh, H., Komori, T., and et al. (1989). Pathogenic significance of interleukin-6 (IL-6/BSF-2) in Castleman's disease. Blood 74, 1360-1367.

Yun, S., Reynolds, R.P., Petrof, I., White, A., Rivera, P.D., Segev, A., Gibson, A.D., Suarez, M., DeSalle, M.J., Ito, N., et al. (2018). Stimulation of entorhinal cortex-dentate gyrus circuitry is antidepressive. Nat Med 24, 658-666. 10.1038/s41591-018-0002-1.

参考文献をもっと見る