Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 Hyaluronic acid has anti-inflammatory properties in intervertebral disc regeneration + 852 word(s) 852 2020-09-16 03:22:03 |
2 format change Meta information modification 852 2020-10-12 03:56:51 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kazezian, Z.; Joyce, K.; Pandit, A. Hyaluronic Acid in Treating IVD. Encyclopedia. Available online: https://encyclopedia.pub/entry/2337 (accessed on 28 March 2024).
Kazezian Z, Joyce K, Pandit A. Hyaluronic Acid in Treating IVD. Encyclopedia. Available at: https://encyclopedia.pub/entry/2337. Accessed March 28, 2024.
Kazezian, Zepur, Kieran Joyce, Abhay Pandit. "Hyaluronic Acid in Treating IVD" Encyclopedia, https://encyclopedia.pub/entry/2337 (accessed March 28, 2024).
Kazezian, Z., Joyce, K., & Pandit, A. (2020, October 04). Hyaluronic Acid in Treating IVD. In Encyclopedia. https://encyclopedia.pub/entry/2337
Kazezian, Zepur, et al. "Hyaluronic Acid in Treating IVD." Encyclopedia. Web. 04 October, 2020.
Hyaluronic Acid in Treating IVD
Edit

High molecular weight hyaluronic acid (HMW HA) and HA-based biomaterials used as therapeutic delivery platforms have been trialled in preclinical models and clinical trials. 

Hyaluronic acid interverebral disc anti-inflammatory

1. Introduction

Hyaluronic acid (HA), which was first isolated in the 1930s, is a distinctive glycosaminoglycan, which, unlike other glycosaminoglycans such as heparan sulphate, chondroitin sulphate, and keratan sulphate, is non-sulphated[1]. It is widely used in treating osteoarthritis[2][3][4][5][6][7][8] and in tissue-engineering approaches for disc repair[9][10]. HA (Figure 3) is an abundant polymer of the ECM with a molecular weight of approximately 106–107 Da in vivo[11]. HA molecule comprises of chains of constantly repeating disaccharides, glucuronic acid and N-acetylglucosamine, which are symbolized by GlcA-β (1→3) and GlcNAc-β (1→4) respectively (Figure 1)[12]. On the intracellular aspect of the cell membrane, synthesis of HA occurs through the hyaluronan synthases (HAS) family, comprised of three types of synthases, activated via different GFs to produce diverse forms of polymerized HA[13]. HAS1 and HAS2 produce the high molecular weight (HMW) of HA of 2–4 × 106 Da, while the low molecular weight of HA (LMW) of 1 × 105–1 × 106 Da is produced by HAS3[13]. These enzymes are regulated by miRNAs[14] and GFs such as TGF-β[15]. Once synthesized, HA is secreted either on the cell surface from the plasma membrane or into the ECM because of its large size[13][16]. The body composition of HA is 15 g[17]. Daily, 33% of HA is turned over through several roots: (i). via the lymphatic system or (ii). via the circulation through the liver or (iii). lysosomal digestion after HA binds to its receptor and is internalized[18]. Furthermore, HA turnover can take place as a result of pathological conditions such as tissue injury and remodelling as a result of oxidative stress and free radicals, HA internalization and degradation, and clearance of HA from tissues via the lymphatics and vascular system[19]. Regarding the receptors through which HA binds to be cleared by the liver and lymph nodes, there are particular receptors such as the hyaluronan receptor for endocytosis 1 (HARE1), lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) and CD44. From the previously mentioned three receptors, HARE1 is considered the major role player through the lymph and vasculature[18]. HA is internalized and degraded when it cannot be removed by the vasculature and lymph[19][20]. In this process, first HA binds to one of its receptors such as receptor for HA-mediated motility (RHAMM), layilin or CD44 after which it is degraded by hyaluronidase-2[15][19][20], then it is digested into fragments through N-glucuronidases and N-acetylglucosaminidases to completely break down the remaining glycans into monosaccharides[20].

Figure 1. The anti-inflammatory role of hyaluronic acid (HA). HA molecule (on the top) is represented according to its international symbols by repeated units of glucuronic acid (GlcA-β1-3) and N-acetylglucosamine (GlcNAc-β1-4) disaccharides. The anti-inflammatory role of HA is mediated through reacting with its several receptors such as CD44 and receptor for HA-mediated motility (RHAMM) preventing the cascades signalled by inflammatory cytokines such as IFNα, TNFα and IL1β through deterring the contact with their receptors.

2. The Key Function of Hyaluronic Acid in Repairing the Intervertebral Disc

The biochemical properties of the HA molecule are entirely dependent on its size[19][21][22]. Within tissues, the HA molecule interacts with various proteins comprising of hyaladherins such as CD44, RHAMM (CD168) (Figure 1). In contrast, inside the ECM, it interacts with N-terminals of proteoglycans such as aggrecan (ACAN), versican, brevican, neurocan and TNFα-stimulated gene-6 (TSG-6) protein[19][23]. These fundamental interactions are essential for HA function in terms of cell-to-cell communication and signalling[15][24]. The mechanical properties of HA can be attributed to its high molecular weight structure. HA is capable of absorbing water approximately 10–104 times its mass as it is negatively charged and is considered an osmotically active molecule[14][15][25][26] . Due to its large volume while hydrated, it can fill large spaces, therefore absorbing shocks and acting like a lubricant[25][27].

HA-CD44 interactions mediate endocytic activity and activate Rho and Rac1 GTPases to regulate cytoskeletal organization[28]. CD44 activation also activates src-related tyrosine kinases to induce cell proliferation through NF-κB signalling[29]. RHAMM activates pp60c-src tyrosine kinase to modulate focal adhesions for RHAMM-mediated cell mobility[30]. Its ability to form large extracellular networks through cell–matrix binding inhibits cytokine binding to prevent downstream signalling (Figure 1)[25][31]. Therefore, HMW HA is capable of inhibiting the signalling of inflammatory cytokines and matrix-degrading enzymes (TNF-α, IL-8, iNOS, aggrecanase 2, matrix metalloproteinases (MMP)s) that were evident upon treating chondrocytes with HMW HA, which also resulted in inhibition of phagocytosis and macrophage activation[32][33]. In tissue injury and remodelling, enzymes break down HMW HA resulting in fragments of LMW HA which can induce phosphorylation of p38 or p42 or p44 mitogen-activated protein kinase (MAPK) and NF-κB which are associated with toll-like receptor (TLR)4 signalling[13][34]. Depending on the tissue type and the size of LMW HA fragments (f-HA)s, pro-inflammatory cytokines can be induced[13][31]; therefore, angiogenesis and tissue remodelling is stimulated[19][21]. It was discovered that human disc cells treated with 6–12 disaccharide fragments of HA can dramatically up-regulate catabolic enzymes such as IL-1β, IL-8, IL-6, MMP1, MMP13, and cyclooxygenase (COX2). Furthermore, it was identified that TLR-2 mediated the role of f-HAs in stimulating the protein expression of IL-6, and both IL-6 and MMP-3 were supported by the MAPK signalling pathway[35].

References

  1. Jiurong Liang; Dianhua Jiang; Paul W. Noble; Hyaluronan as a therapeutic target in human diseases. Advanced Drug Delivery Reviews 2016, 97, 186-203, 10.1016/j.addr.2015.10.017.
  2. Steven Bowman; Mohamed E. Awad; Mark W. Hamrick; Monte Hunter; Sadanand Fulzele; Recent advances in hyaluronic acid based therapy for osteoarthritis. Clinical and Translational Medicine 2018, 7, 1-11, 10.1186/s40169-017-0180-3.
  3. Lai, H.Y.; Chen, Y.C.; Chen, T.J.; Chou, L.F.; Chen, L.K.; Hwang, S.J. Intra-articular hyaluronic acid for treatment of osteoarthritis: A nationwide study among the older population of Taiwan. BMC Health Serv. Res. 2008, 8, 24.
  4. Chou, C.L.; Li, H.W.; Lee, S.H.; Tsai, K.L.; Ling, H.Y. Effect of intra-articular injection of hyaluronic acid in rheumatoid arthritis patients with knee osteoarthritis. J. Chin. Med. Assoc. 2008, 71, 411–415.
  5. Migliore, A.; Granata, M. Intra-articular use of hyaluronic acid in the treatment of osteoarthritis. Clin. Interv. Aging 2008, 3, 365–369.
  6. Chircov, C.; Grumezescu, A.M.; Bejenaru, L.E. Hyaluronic acid-based scaffolds for tissue engineering. Rom. J. Morphol. Embryol. 2018, 59, 71–76.
  7. Cooper, C.; Rannou, F.; Richette, P.; Bruyere, O.; Al-Daghri, N.; Altman, R.D.; Brandi, M.L.; Collaud Basset, S.; Herrero-Beaumont, G.; Migliore, A.; et al. Use of intraarticular hyaluronic acid in the management of knee osteoarthritis in clinical practice. Arthritis Care Res. 2017, 69, 1287–1296.
  8. Stirma, G.A.; Chaves, D.H.; Tortato, S.; Belangero, P.S.; Lara, P.H.S.; Ejnisman, B. Prospective evaluation of periarticular hyaluronic acid infiltration for the treatment of lateral epicondylitis. Acta Ortop. Bras. 2020, 28, 107–110
  9. Isa, I.L.; Srivastava, A.; Tiernan, D.; Owens, P.; Rooney, P.; Dockery, P.; Pandit, A. Hyaluronic acid based hydrogels attenuate inflammatory receptors and neurotrophins in interleukin-1beta induced inflammation model of nucleus pulposus cells. Biomacromolecules 2015, 16, 1714–1725.
  10. Kazezian, Z.; Li, Z.; Alini, M.; Grad, S.; Pandit, A. Injectable hyaluronic acid down-regulates interferon signaling molecules, IGFBP3 and IFIT3 in the bovine intervertebral disc. Acta Biomater. 2017, 52, 118–129
  11. Toshiyuki Emurai; Lipid Raft-Mediated Regulation of Hyaluronan–CD44 Interactions in Inflammation and Cancer. Frontiers in Immunology 2015, 6, 420, 10.3389/fimmu.2015.00420.
  12. Bryan P. Toole; Hyaluronan in morphogenesis. Journal of Internal Medicine 1997, 242, 35-40, 10.1046/j.1365-2796.1997.00171.x.
  13. Larry S Sherman; Neural stem cell niches Roles for the hyaluronan-based extracellular matrix. Frontiers in Bioscience 2011, 3, 1165-1179, 10.2741/s218.
  14. Anne Karine Lagendijk; András Szabó; Roeland M.H. Merks; Jeroen Bakkers; Hyaluronan: A critical regulator of endothelial-to-mesenchymal transition during cardiac valve formation. Trends in Cardiovascular Medicine 2013, 23, 135-142, 10.1016/j.tcm.2012.10.002.
  15. E. R. Bastow; Sharon Byers; S. B. Golub; C. E. Clarkin; A. A. Pitsillides; Amanda J. Fosang; Hyaluronan synthesis and degradation in cartilage and bone. Cellular and Molecular Life Sciences 2007, 65, 395-413, 10.1007/s00018-007-7360-z.
  16. F Bonnet; D G Dunham; Tim E Hardingham; Structure and interactions of cartilage proteoglycan binding region and link protein. Biochemical Journal 1985, 228, 77-85, 10.1042/bj2280077.
  17. Amir Fakhari; Cory J. Berkland; Applications and emerging trends of hyaluronic acid in tissue engineering, as a dermal filler and in osteoarthritis treatment.. Acta Biomaterialia 2013, 9, 7081-7092, 10.1016/j.actbio.2013.03.005.
  18. Hascall, V.; Esko, J.D. Hyaluronan. In Essentials of Glycobiology, 2nd ed.; Varki, A., Cummings, R.D., Esko, J.D., Freeze, H.H., Stanley, P., Bertozzi, C.R., Hart, G.W., Etzler, M.E., Eds.; Cold Spring Harbor (NY): New York, NY, USA, 2009.
  19. Robert Stern; Akira A. Asari; Kazuki N. Sugahara; Hyaluronan fragments: An information-rich system. European Journal of Cell Biology 2006, 85, 699-715, 10.1016/j.ejcb.2006.05.009.
  20. Pouria Moshayedi; S. Thomas Carmichael; Hyaluronan, neural stem cells and tissue reconstruction after acute ischemic stroke. Biomatter 2013, 3, e23863, 10.4161/biom.23863.
  21. Stern, R.; Kogan, G.; Jedrzejas, M.J.; Soltes, L. The many ways to cleave hyaluronan. Biotech. Adv. 2007, 25, 537–557.
  22. Safrankova, B.; Gajdova, S.; Kubala, L. The potency of hyaluronan of different molecular weights in the stimulation of blood phagocytes. Mediat. Inflamm. 2010, 2010, 380948.
  23. Durba Mukhopadhyay; Akira Asari; Marilyn S. Rugg; Anthony J Day; Csaba Fülöp; Specificity of the Tumor Necrosis Factor-induced Protein 6-mediated Heavy Chain Transfer from Inter-α-trypsin Inhibitor to Hyaluronan. Journal of Biological Chemistry 2004, 279, 11119-11128, 10.1074/jbc.m313471200.
  24. Bryan P Toole; Hyaluronan in morphogenesis. Seminars in Cell & Developmental Biology 2001, 12, 79-87, 10.1006/scdb.2000.0244.
  25. Toole, B.P. Hyaluronan: From extracellular glue to pericellular cue. Nat. Rev. Cancer 2004, 4, 528–539.
  26. Collins, M.N.; Birkinshaw, C. Hyaluronic acid based scaffolds for tissue engineering—A review. Carb Polym. 2013, 92, 1262–1279.
  27. Eric J. Strauss; Jennifer A. Hart; Mark D. Miller; Roy D. Altman; Jeffrey Rosen; Hyaluronic Acid Viscosupplementation and Osteoarthritis. The American Journal of Sports Medicine 2009, 37, 1636-1644, 10.1177/0363546508326984.
  28. Eva A. Turley; Paul W. Noble; Lilly Y. W. Bourguignon; Signaling Properties of Hyaluronan Receptors. Journal of Biological Chemistry 2001, 277, 4589-4592, 10.1074/jbc.r100038200.
  29. Katherine A. Fitzgerald; Andrew G. Bowie; Barbara Sheehy Skeffington; Luke A. J. O’Neill; Ras, Protein Kinase Cζ, and IκB Kinases 1 and 2 Are Downstream Effectors of CD44 During the Activation of NF-κB by Hyaluronic Acid Fragments in T-24 Carcinoma Cells. The Journal of Immunology 2000, 164, 2053-2063, 10.4049/jimmunol.164.4.2053.
  30. C L Hall; E.A Turley; Hyaluronan: RHAMM mediated cell locomotion and signaling in tumorigenesis.. Journal of Neuro-Oncology 1995, 26, 221-229.
  31. Paul L. Bollyky.; Marika Bogdani; Jennifer B. Bollyky; Rebecca L. Hull; Thomas N. Wight; The Role of Hyaluronan and the Extracellular Matrix in Islet Inflammation and Immune Regulation. Current Diabetes Reports 2012, 12, 471-480, 10.1007/s11892-012-0297-0.
  32. Wang, Q.G.; El Haj, A.J.; Kuiper, N.J. Glycosaminoglycans in the pericellular matrix of chondrons and chondrocytes. J. Anat. 2008, 213, 266–273.
  33. Pauloin, T.; Dutot, M.; Warnet, J.M.; Rat, P. In vitro modulation of preservative toxicity: High molecular weight hyaluronan decreases apoptosis and oxidative stress induced by benzalkonium chloride. Eur. J. Pharm. Sci. 2008, 34, 263–273.
  34. Kristen R. Taylor; Janet M. Trowbridge; Jennifer A. Rudisill; Christian C. Termeer; Jan C. Simon; Richard L. Gallo; Hyaluronan Fragments Stimulate Endothelial Recognition of Injury through TLR4. Journal of Biological Chemistry 2004, 279, 17079-17084, 10.1074/jbc.m310859200.
  35. Lilian Quero; Marina Klawitter; Anja Schmaus; Melanie Rothley; Jonathan P. Sleeman; André Tiaden; Juergen Klasen; Norbert Boos; Michael O. Hottiger; Karin Wuertz; et al.Peter J. Richards Hyaluronic acid fragments enhance the inflammatory and catabolic response in human intervertebral disc cells through modulation of toll-like receptor 2 signalling pathways. Arthritis Research & Therapy 2013, 15, R94-R94, 10.1186/ar4274.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 939
Revisions: 2 times (View History)
Update Date: 12 Oct 2020
1000/1000