Anti-Inflammatory Therapy for Temporomandibular Joint Osteoarthritis Using mRNA Medicine Encoding Interleukin-1 Receptor Antagonist
Abstract
:1. Introduction
2. Materials and Methods
2.1. Chemicals
2.2. Synthesis of Block Copolymers
2.3. Preparation of mRNA
2.4. Preparation of mRNA-Loaded Polyplex Nanomicelles
2.5. Animal Model
2.6. Pain Behavior Assessment
2.7. Micro-Computed Tomography Evaluation
2.8. Histological and Immunofluorescence Analyses
2.9. Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR)
2.10. Western Blot
2.11. Statistical Analysis
3. Results
3.1. Evaluation of mRNA Delivery into the Articular Cartilage
3.2. IL-1Ra mRNA Continuously Suppresses Pain from an Early Time Point
3.3. IL-1Ra mRNA Alleviates Degeneration of Cartilage and Bone
3.4. IL-1Ra mRNA Modulates OA-Induced Inflammation
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Acknowledgments
Conflicts of Interest
References
- Zarb, G.A.; Carlsson, G.E. Temporomandibular disorders: Osteoarthritis. J. Orofac. Pain 1999, 13, 295–306. [Google Scholar] [PubMed]
- Stegenga, B.; de Bont, L.G.; Boering, G. Osteoarthrosis as the cause of craniomandibular pain and dysfunction: A unifying concept. J. Oral Maxillofac. Surg. 1989, 47, 249–256. [Google Scholar] [CrossRef]
- Wang, X.D.; Zhang, J.N.; Gan, Y.H.; Zhou, Y.H. Current understanding of pathogenesis and treatment of TMJ osteoarthritis. J. Dent. Res. 2015, 94, 666–673. [Google Scholar] [CrossRef]
- Vernal, R.; Velásquez, E.; Gamonal, J.; Garcia-Sanz, J.A.; Silva, A.; Sanz, M. Expression of proinflammatory cytokines in osteoarthritis of the temporomandibular joint. Arch. Oral Biol. 2008, 53, 910–915. [Google Scholar] [CrossRef]
- Ning, L.; Ishijima, M.; Kaneko, H.; Kurihara, H.; Arikawa-Hirasawa, E.; Kubota, M.; Liu, L.; Xu, Z.; Futami, I.; Yusup, A.; et al. Correlations between both the expression levels of inflammatory mediators and growth factor in medial perimeniscal synovial tissue and the severity of medial knee osteoarthritis. Int. Orthop. 2011, 35, 831–838. [Google Scholar] [CrossRef]
- Takahashi, T.; Kondoh, T.; Fukuda, M.; Yamazaki, Y.; Toyosaki, T.; Suzuki, R. Proinflammatory cytokines detectable in synovial fluids from patients with temporomandibular disorders. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. ENDOD. 1998, 85, 135–141. [Google Scholar] [CrossRef]
- Winzen, R.; Kracht, M.; Ritter, B.; Wilhelm, A.; Chen, C.Y.; Shyu, A.B.; Müller, M.; Gaestel, M.; Resch, K.; Holtmann, H. The p38 MAP kinase pathway signals for cytokine-induced mRNA stabilization via MAP kinase-activated protein kinase 2 and an AU-rich region-targeted mechanism. EMBO J. 1999, 18, 4969–4980. [Google Scholar] [CrossRef]
- Mengshol, J.A.; Vincenti, M.P.; Brinckerhoff, C.E. IL-1 induces collagenase-3 (MMP-13) promoter activity in stably transfected chondrocytic cells: Requirement for Runx-2 and activation by p38 MAPK and JNK pathways. Nucleic Acids Res. 2001, 29, 4361–4372. [Google Scholar] [CrossRef]
- Tetlow, L.C.; Adlam, D.J.; Woolley, D.E. Matrix metalloproteinase and proinflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: Associations with degenerative changes. Arthritis Rheum. 2001, 44, 585–594. [Google Scholar] [CrossRef]
- Mantovani, A.; Dinarello, C.A.; Molgora, M.; Garlanda, C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019, 50, 778–795. [Google Scholar] [CrossRef] [Green Version]
- Caron, J.P.; Fernandes, J.C.; Martel-Pelletier, J.; Tardif, G.; Mineau, F.; Geng, C.; Pelletier, J.P. Chondroprotective effect of intraarticular injections of interleukin-1 receptor antagonist in experimental osteoarthritis. Suppression of collagenase-1 expression. Arthritis Rheum. 1996, 39, 1535–1544. [Google Scholar] [CrossRef] [PubMed]
- Elsaid, K.A.; Zhang, L.; Shaman, Z.; Patel, C.; Schmidt, T.A.; Jay, G.D. The impact of early intra-articular administration of interleukin-1 receptor antagonist on lubricin metabolism and cartilage degeneration in an anterior cruciate ligament transection model. Osteoarthr. Cartil. 2015, 23, 114–121. [Google Scholar] [CrossRef] [PubMed]
- Eloseily, E.M.; Weiser, P.; Crayne, C.B.; Haines, H.; Mannion, M.L.; Stoll, M.L.; Beukelman, T.; Atkinson, T.P.; Cron, R.Q. Benefit of Anakinra in Treating Pediatric Secondary Hemophagocytic Lymphohistiocytosis. Arthritis Rheumatol. 2020, 72, 326–334. [Google Scholar] [CrossRef]
- Chevalier, X.; Goupille, P.; Beaulieu, A.D.; Burch, F.X.; Bensen, W.G.; Conrozier, T.; Loeuille, D.; Kivitz, A.J.; Silver, D.; Appleton, B.E. Intraarticular injection of anakinra in osteoarthritis of the knee: A multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum. 2009, 61, 344–352. [Google Scholar] [CrossRef]
- Cohen, S.; Hurd, E.; Cush, J.; Schiff, M.; Weinblatt, M.E.; Moreland, L.W.; Kremer, J.; Bear, M.B.; Rich, W.J.; McCabe, D. Treatment of rheumatoid arthritis with anakinra, a recombinant human interleukin-1 receptor antagonist, in combination with methotrexate: Results of a twenty-four-week, multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2002, 46, 614–624. [Google Scholar] [CrossRef]
- Larsen, C.M.; Faulenbach, M.; Vaag, A.; Vølund, A.; Ehses, J.A.; Seifert, B.; Mandrup-Poulsen, T.; Donath, M.Y. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N. Engl. J. Med. 2007, 356, 1517–1526. [Google Scholar] [CrossRef]
- Wangerek, L.A.; Dahl, H.H.; Senden, T.J.; Carlin, J.B.; Jans, D.A.; Dunstan, D.E.; Ioannou, P.A.; Williamson, R.; Forrest, S.M. Atomic force microscopy imaging of DNA-cationic liposome complexes optimised for gene transfection into neuronal cells. J. Gene Med. 2001, 3, 72–81. [Google Scholar] [CrossRef]
- Zou, S.; Scarfo, K.; Nantz, M.H.; Hecker, J.G. Lipid-mediated delivery of RNA is more efficient than delivery of DNA in non-dividing cells. Int. J. Pharm. 2010, 389, 232–243. [Google Scholar] [CrossRef] [PubMed]
- Sahin, U.; Kariko, K.; Tureci, O. mRNA-based therapeutics--developing a new class of drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780. [Google Scholar] [CrossRef]
- Warren, L.; Manos, P.D.; Ahfeldt, T.; Loh, Y.H.; Li, H.; Lau, F.; Ebina, W.; Mandal, P.K.; Smith, Z.D.; Meissner, A.; et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 2010, 7, 618–630. [Google Scholar] [CrossRef] [Green Version]
- Tavernier, G.; Wolfrum, K.; Demeester, J.; De Smedt, S.C.; Adjaye, J.; Rejman, J. Activation of pluripotency-associated genes in mouse embryonic fibroblasts by non-viral transfection with in vitro-derived mRNAs encoding Oct4, Sox2, Klf4 and cMyc. Biomaterials 2012, 33, 412–417. [Google Scholar] [CrossRef] [PubMed]
- Kedmi, R.; Ben-Arie, N.; Peer, D. The systemic toxicity of positively charged lipid nanoparticles and the role of Toll-like receptor 4 in immune activation. Biomaterials 2010, 31, 6867–6875. [Google Scholar] [CrossRef] [PubMed]
- Sedic, M.; Senn, J.J.; Lynn, A.; Laska, M.; Smith, M.; Platz, S.J.; Bolen, J.; Hoge, S.; Bulychev, A.; Jacquinet, E.; et al. Safety Evaluation of Lipid Nanoparticle-Formulated Modified mRNA in the Sprague-Dawley Rat and Cynomolgus Monkey. Vet. Pathol. 2018, 55, 341–354. [Google Scholar] [CrossRef] [PubMed]
- Kanayama, N.; Fukushima, S.; Nishiyama, N.; Itaka, K.; Jang, W.D.; Miyata, K.; Yamasaki, Y.; Chung, U.I.; Kataoka, K. A PEG-based biocompatible block catiomer with high buffering capacity for the construction of polyplex micelles showing efficient gene transfer toward primary cells. ChemMedChem 2006, 1, 439–444. [Google Scholar] [CrossRef]
- Uchida, H.; Miyata, K.; Oba, M.; Ishii, T.; Suma, T.; Itaka, K.; Nishiyama, N.; Kataoka, K. Odd-even effect of repeating aminoethylene units in the side chain of N-substituted polyaspartamides on gene transfection profiles. J. Am. Chem. Soc. 2011, 133, 15524–15532. [Google Scholar] [CrossRef]
- Uchida, H.; Itaka, K.; Nomoto, T.; Ishii, T.; Suma, T.; Ikegami, M.; Miyata, K.; Oba, M.; Nishiyama, N.; Kataoka, K. Modulated protonation of side chain aminoethylene repeats in N-substituted polyaspartamides promotes mRNA transfection. J. Am. Chem. Soc. 2014, 136, 12396–12405. [Google Scholar] [CrossRef]
- Itaka, K.; Kataoka, K. Progress and prospects of polyplex nanomicelles for plasmid DNA delivery. Curr. Gene Ther. 2011, 11, 457–465. [Google Scholar] [CrossRef]
- Uchida, S.; Itaka, K.; Uchida, H.; Hayakawa, K.; Ogata, T.; Ishii, T.; Fukushima, S.; Osada, K.; Kataoka, K. In vivo messenger RNA introduction into the central nervous system using polyplex nanomicelle. PLoS ONE 2013, 8, e56220. [Google Scholar] [CrossRef]
- Matsui, A.; Uchida, S.; Ishii, T.; Itaka, K.; Kataoka, K. Messenger RNA-based therapeutics for the treatment of apoptosis-associated diseases. Sci. Rep. 2015, 5, 15810. [Google Scholar] [CrossRef]
- Aini, H.; Itaka, K.; Fujisawa, A.; Uchida, H.; Uchida, S.; Fukushima, S.; Kataoka, K.; Saito, T.; Chung, U.I.; Ohba, S. Messenger RNA delivery of a cartilage-anabolic transcription factor as a disease-modifying strategy for osteoarthritis treatment. Sci. Rep. 2016, 6, 18743. [Google Scholar] [CrossRef] [Green Version]
- Lin, C.Y.; Perche, F.; Ikegami, M.; Uchida, S.; Kataoka, K.; Itaka, K. Messenger RNA-based therapeutics for brain diseases: An animal study for augmenting clearance of beta-amyloid by intracerebral administration of neprilysin mRNA loaded in polyplex nanomicelles. J. Control. Release 2016, 235, 268–275. [Google Scholar] [CrossRef] [PubMed]
- Crowley, S.T.; Fukushima, Y.; Uchida, S.; Kataoka, K.; Itaka, K. Enhancement of Motor Function Recovery after Spinal Cord Injury in Mice by Delivery of Brain-Derived Neurotrophic Factor mRNA. Mol. Ther. Nucleic Acids 2019, 17, 465–476. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.Y.; Crowley, S.T.; Uchida, S.; Komaki, Y.; Kataoka, K.; Itaka, K. Treatment of Intervertebral Disk Disease by the Administration of mRNA Encoding a Cartilage-Anabolic Transcription Factor. Mol. Ther. Nucleic Acids 2019, 16, 162–171. [Google Scholar] [CrossRef]
- Fukushima, Y.; Uchida, S.; Imai, H.; Nakatomi, H.; Kataoka, K.; Saito, N.; Itaka, K. Treatment of ischemic neuronal death by introducing brain-derived neurotrophic factor mRNA using polyplex nanomicelle. Biomaterials 2021, 270, 120681. [Google Scholar] [CrossRef]
- Oyama, N.; Kawaguchi, M.; Itaka, K.; Kawakami, S. Efficient Messenger RNA Delivery to the Kidney Using Renal Pelvis Injection in Mice. Pharmaceutics 2021, 13, 1810. [Google Scholar] [CrossRef]
- Hashimoto, Y.; Kuniishi, H.; Sakai, K.; Fukushima, Y.; Du, X.; Yamashiro, K.; Hori, K.; Imamura, M.; Hoshino, M.; Yamada, M.; et al. Brain Dp140 alters glutamatergic transmission and social behaviour in the mdx52 mouse model of Duchenne muscular dystrophy. Prog. Neurobiol. 2022, 216, 102288. [Google Scholar] [CrossRef]
- Wang, X.D.; Kou, X.X.; He, D.Q.; Zeng, M.M.; Meng, Z.; Bi, R.Y.; Liu, Y.; Zhang, J.N.; Gan, Y.H.; Zhou, Y.H. Progression of cartilage degradation, bone resorption and pain in rat temporomandibular joint osteoarthritis induced by injection of iodoacetate. PLoS ONE 2012, 7, e45036. [Google Scholar] [CrossRef]
- Ren, K. An improved method for assessing mechanical allodynia in the rat. Physiol. Behav. 1999, 67, 711–716. [Google Scholar] [CrossRef]
- Kawamoto, T.; Kawamoto, K. Preparation of thin frozen sections from nonfixed and undecalcified hard tissues using Kawamot’s film method (2012). Methods Mol. Biol. 2014, 1130, 149–164. [Google Scholar] [CrossRef]
- Mankin, H.J.; Dorfman, H.; Lippiello, L.; Zarins, A. Biochemical and metabolic abnormalities in articular cartilage from osteo-arthritic human hips. II. Correlation of morphology with biochemical and metabolic data. J. Bone Jt. Surg. Am. 1971, 53, 523–537. [Google Scholar] [CrossRef]
- Pauli, C.; Whiteside, R.; Heras, F.L.; Nesic, D.; Koziol, J.; Grogan, S.P.; Matyas, J.; Pritzker, K.P.; D’Lima, D.D.; Lotz, M.K. Comparison of cartilage histopathology assessment systems on human knee joints at all stages of osteoarthritis development. Osteoarthr. Cartil. 2012, 20, 476–485. [Google Scholar] [CrossRef] [PubMed]
- Peeters, M.; Huang, C.L.; Vonk, L.A.; Lu, Z.F.; Bank, R.A.; Helder, M.N.; Doulabi, B.Z. Optimisation of high-quality total ribonucleic acid isolation from cartilaginous tissues for real-time polymerase chain reaction analysis. Bone Jt. Res. 2016, 5, 560–568. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Teo, K.Y.W.; Chuah, S.J.; Lai, R.C.; Lim, S.K.; Toh, W.S. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials 2019, 200, 35–47. [Google Scholar] [CrossRef]
- Kim, H.; Yang, G.; Park, J.; Choi, J.; Kang, E.; Lee, B.K. Therapeutic effect of mesenchymal stem cells derived from human umbilical cord in rabbit temporomandibular joint model of osteoarthritis. Sci. Rep. 2019, 9, 13854. [Google Scholar] [CrossRef]
- Jiang, H.; Xu, L.; Liu, W.; Xiao, M.; Ke, J.; Long, X. Chronic Pain Causes Peripheral and Central Responses in MIA-Induced TMJOA Rats. Cell. Mol. Neurobiol. 2021, 42, 1441–1451. [Google Scholar] [CrossRef]
- Hou, X.; Zaks, T.; Langer, R.; Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 2021, 6, 1078–1094. [Google Scholar] [CrossRef] [PubMed]
- Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416. [Google Scholar] [CrossRef]
- Vergnes, J.N. Safety and Efficacy of the BNT162b2 mRNA COVID-19 Vaccine. N. Engl. J. Med. 2021, 384, 1576–1578. [Google Scholar] [CrossRef]
- Maugeri, M.; Nawaz, M.; Papadimitriou, A.; Angerfors, A.; Camponeschi, A.; Na, M.; Hölttä, M.; Skantze, P.; Johansson, S.; Sundqvist, M.; et al. Linkage between endosomal escape of LNP-mRNA and loading into EVs for transport to other cells. Nat. Commun. 2019, 10, 4333. [Google Scholar] [CrossRef]
- Karikó, K.; Buckstein, M.; Ni, H.; Weissman, D. Suppression of RNA recognition by Toll-like receptors: The impact of nucleoside modification and the evolutionary origin of RNA. Immunity 2005, 23, 165–175. [Google Scholar] [CrossRef] [Green Version]
- Ji, R.R.; Xu, Z.Z.; Gao, Y.J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 2014, 13, 533–548. [Google Scholar] [CrossRef] [PubMed]
- Woolf, C.J. Evidence for a central component of post-injury pain hypersensitivity. Nature 1983, 306, 686–688. [Google Scholar] [CrossRef] [PubMed]
- Aley, K.O.; Levine, J.D. Role of protein kinase A in the maintenance of inflammatory pain. J. Neurosci. 1999, 19, 2181–2186. [Google Scholar] [CrossRef]
- Gold, M.S.; Levine, J.D.; Correa, A.M. Modulation of TTX-R INa by PKC and PKA and their role in PGE2-induced sensitization of rat sensory neurons in vitro. J. Neurosci. 1998, 18, 10345–10355. [Google Scholar] [CrossRef] [PubMed]
- Costigan, M.; Moss, A.; Latremoliere, A.; Johnston, C.; Verma-Gandhu, M.; Herbert, T.A.; Barrett, L.; Brenner, G.J.; Vardeh, D.; Woolf, C.J.; et al. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J. Neurosci. 2009, 29, 14415–14422. [Google Scholar] [CrossRef] [PubMed]
- Kawasaki, Y.; Zhang, L.; Cheng, J.K.; Ji, R.R. Cytokine mechanisms of central sensitization: Distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J. Neurosci. 2008, 28, 5189–5194. [Google Scholar] [CrossRef]
- Bove, S.E.; Calcaterra, S.L.; Brooker, R.M.; Huber, C.M.; Guzman, R.E.; Juneau, P.L.; Schrier, D.J.; Kilgore, K.S. Weight bearing as a measure of disease progression and efficacy of anti-inflammatory compounds in a model of monosodium iodoacetate-induced osteoarthritis. Osteoarthr. Cartil. 2003, 11, 821–830. [Google Scholar] [CrossRef]
- Berenbaum, F. Signaling transduction: Target in osteoarthritis. Curr. Opin. Rheumatol. 2004, 16, 616–622. [Google Scholar] [CrossRef]
- Blanco, E.; Shen, H.; Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 2015, 33, 941–951. [Google Scholar] [CrossRef]
- Semple, S.C.; Akinc, A.; Chen, J.; Sandhu, A.P.; Mui, B.L.; Cho, C.K.; Sah, D.W.; Stebbing, D.; Crosley, E.J.; Yaworski, E.; et al. Rational design of cationic lipids for siRNA delivery. Nat. Biotechnol. 2010, 28, 172–176. [Google Scholar] [CrossRef]
- Kim, J.; Eygeris, Y.; Gupta, M.; Sahay, G. Self-assembled mRNA vaccines. Adv. Drug Deliv. Rev. 2021, 170, 83–112. [Google Scholar] [CrossRef] [PubMed]
- Yanez Arteta, M.; Kjellman, T.; Bartesaghi, S.; Wallin, S.; Wu, X.; Kvist, A.J.; Dabkowska, A.; Székely, N.; Radulescu, A.; Bergenholtz, J.; et al. Successful reprogramming of cellular protein production through mRNA delivered by functionalized lipid nanoparticles. Proc. Natl. Acad. Sci. USA 2018, 115, E3351–E3360. [Google Scholar] [CrossRef] [PubMed]
- Kulkarni, J.A.; Witzigmann, D.; Leung, J.; van der Meel, R.; Zaifman, J.; Darjuan, M.M.; Grisch-Chan, H.M.; Thöny, B.; Tam, Y.Y.C.; Cullis, P.R. Fusion-dependent formation of lipid nanoparticles containing macromolecular payloads. Nanoscale 2019, 11, 9023–9031. [Google Scholar] [CrossRef] [PubMed]
- Heyes, J.; Hall, K.; Tailor, V.; Lenz, R.; MacLachlan, I. Synthesis and characterization of novel poly(ethylene glycol)-lipid conjugates suitable for use in drug delivery. J. Control. Release 2006, 112, 280–290. [Google Scholar] [CrossRef] [PubMed]
- Pardi, N.; Tuyishime, S.; Muramatsu, H.; Kariko, K.; Mui, B.L.; Tam, Y.K.; Madden, T.D.; Hope, M.J.; Weissman, D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J. Control. Release 2015, 217, 345–351. [Google Scholar] [CrossRef] [Green Version]
Gene | Forward | Reverse |
---|---|---|
Human | ||
IL-1Ra | 5′-AACCTTCTACCTGCGGAACA-3′ | 5′-GCCAGACTTCACACAGCTCA-3′ |
Rat | ||
β-Actin | 5′-CACCCGCGAGTACAACCTTCT-3′ | 5′-TCGTCATCCATGGCGAACTGG-3′ |
IL-1β | 5′-TGTCTGACCCATGTGAGCTG-3′ | 5′-TTTGGGATCCACACTCTCCA -3′ |
IL-6 | 5′-GTTTCTCTCCGCAAGAGACTTC -3′ | 5′-TGTGGGTGGTATCCTCTGTGA-3′ |
TNF-α | 5′-ACTGAACTTCGGGGTGATCG-3′ | 5′-TCCGCTTGGTGGTTTGCTAC-3′ |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Deng, J.; Fukushima, Y.; Nozaki, K.; Nakanishi, H.; Yada, E.; Terai, Y.; Fueki, K.; Itaka, K. Anti-Inflammatory Therapy for Temporomandibular Joint Osteoarthritis Using mRNA Medicine Encoding Interleukin-1 Receptor Antagonist. Pharmaceutics 2022, 14, 1785. https://doi.org/10.3390/pharmaceutics14091785
Deng J, Fukushima Y, Nozaki K, Nakanishi H, Yada E, Terai Y, Fueki K, Itaka K. Anti-Inflammatory Therapy for Temporomandibular Joint Osteoarthritis Using mRNA Medicine Encoding Interleukin-1 Receptor Antagonist. Pharmaceutics. 2022; 14(9):1785. https://doi.org/10.3390/pharmaceutics14091785
Chicago/Turabian StyleDeng, Jia, Yuta Fukushima, Kosuke Nozaki, Hideyuki Nakanishi, Erica Yada, Yuki Terai, Kenji Fueki, and Keiji Itaka. 2022. "Anti-Inflammatory Therapy for Temporomandibular Joint Osteoarthritis Using mRNA Medicine Encoding Interleukin-1 Receptor Antagonist" Pharmaceutics 14, no. 9: 1785. https://doi.org/10.3390/pharmaceutics14091785
APA StyleDeng, J., Fukushima, Y., Nozaki, K., Nakanishi, H., Yada, E., Terai, Y., Fueki, K., & Itaka, K. (2022). Anti-Inflammatory Therapy for Temporomandibular Joint Osteoarthritis Using mRNA Medicine Encoding Interleukin-1 Receptor Antagonist. Pharmaceutics, 14(9), 1785. https://doi.org/10.3390/pharmaceutics14091785