Abstract
The hyaluronan (HA) matrix in the tissue microenvironment is crucial for maintaining homeostasis by regulating inflammatory signalling, endothelial–mesenchymal transition and cell migration. During development, covalent modifications and osmotic swelling of HA create mechanical forces that initiate midgut rotation, vascular patterning and branching morphogenesis. Together with its main cell surface receptor, CD44, HA establishes a physicochemical scaffold at the cell surface that facilitates the interaction and clustering of growth factors and receptors that is required for normal physiology. High-molecular-weight HA, tumour necrosis factor-stimulated gene 6, pentraxin 3 and CD44 form a stable pericellular matrix that promotes tissue regeneration and reduces inflammation. By contrast, breakdown of high-molecular-weight HA into depolymerized fragments by hyaluronidases triggers inflammatory signalling, leukocyte migration and angiogenesis, contributing to tissue damage and fibrosis in kidney disease. Targeting HA metabolism is challenging owing to its dynamic regulation and tissue-specific functions. Nonetheless, modulating HA matrix functions by targeting its binding partners holds promise as a therapeutic strategy for restoring tissue homeostasis and mitigating pathological processes. Further research in this area is warranted to enable the development of novel therapeutic approaches for kidney and other diseases characterized by dysregulated HA metabolism.
Key points
-
Hyaluronan (HA) is a key component of the extracellular matrix that has critical roles in tissue homeostasis, organogenesis, cell signalling, cell migration and regulation of immune responses.
-
Together with its cell surface receptor CD44 and HA-binding proteins such as TSG6 and pentraxin3, high-molecular-weight (HMW)-HA creates a stable pericellular matrix that facilitates tissue regeneration and reduces inflammation.
-
Depolymerization of HMW-HA enables tissue remodelling and generates HA fragments that can activate inflammatory signalling, recruit leukocytes and promote angiogenesis.
-
During development, covalent modifications and osmotic swelling of HA create mechanical forces that initiate midgut rotation, vascular patterning and branching morphogenesis.
-
In kidney injury and disease, synthesis of HMW-HA contributes to tissue regeneration, whereas dysregulated breakdown of HA has been associated with inflammation, hyalinosis and fibrosis; in renal cell carcinoma, high tumour levels of HA are associated with an unfavourable prognosis.
-
HA metabolism is an unlikely therapeutic target owing to its dynamic, tissue-specific regulation and fundamental importance for tissue homeostasis; however, HA binding partners could potentially be targeted to modulate HA matrix functions and mitigate pathological processes in kidney disease.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Lake, B. B. et al. An atlas of healthy and injured cell states and niches in the human kidney. Nature 619, 585–594 (2023).
Wang, G. et al. Analyzing cell-type-specific dynamics of metabolism in kidney repair. Nat. Metab. 4, 1109–1118 (2022).
Hansell, P. & Palm, F. A role for the extracellular matrix component hyaluronan in kidney dysfunction during ACE-inhibitor fetopathy. Acta Physiol. 213, 795–804 (2015).
Spicer, A. P., Olson, J. S. & McDonald, J. A. Molecular cloning and characterization of a cDNA encoding the third putative mammalian hyaluronan synthase. J. Biol. Chem. 272, 8957–8961 (1997).
Shyjan, A. M., Heldin, P., Butcher, E. C., Yoshino, T. & Briskin, M. J. Functional cloning of the cDNA for a human hyaluronan synthase. J. Biol. Chem. 271, 23395–23399 (1996).
Itano, N. & Kimata, K. Molecular cloning of human hyaluronan synthase. Biochem. Biophys. Res. Commun. 222, 816–820 (1996).
Rilla, K. et al. Plasma membrane residence of hyaluronan synthase is coupled to its enzymatic activity. J. Biol. Chem. 280, 31890–31897 (2005).
Rilla, K. et al. Changed lamellipodial extension, adhesion plaques and migration in epidermal keratinocytes containing constitutively expressed sense and antisense hyaluronan synthase 2 (Has2) genes. J. Cell Sci. 115, 3633–3643 (2002).
Tlapak-Simmons, V. L., Baggenstoss, B. A., Kumari, K., Heldermon, C. & Weigel, P. H. Kinetic characterization of the recombinant hyaluronan synthases from Streptococcus pyogenes and Streptococcus equisimilis. J. Biol. Chem. 274, 4246–4253 (1999).
Itano, N. & Kimata, K. Mammalian hyaluronan synthases. IUBMB Life 54, 195–199 (2002).
Bart, G. et al. Fluorescence resonance energy transfer (FRET) and proximity ligation assays reveal functionally relevant homo- and heteromeric complexes among hyaluronan synthases HAS1, HAS2 and HAS3. J. Biol. Chem. 290, 11479–11490 (2015).
Siiskonen, H., Oikari, S., Pasonen-Seppanen, S. & Rilla, K. Hyaluronan synthase 1: a mysterious enzyme with unexpected functions. Front. Immunol. 6, 43 (2015).
Rilla, K. et al. Hyaluronan synthase 1 (HAS1) requires higher cellular UDP-GlcNAc concentration than HAS2 and HAS3. J. Biol. Chem. 288, 5973–5983 (2013).
Itano, N. et al. Three isoforms of mammalian hyaluronan synthases have distinct enzymatic properties. J. Biol. Chem. 274, 25085–25092 (1999).
Torronen, K. et al. Tissue distribution and subcellular localization of hyaluronan synthase isoenzymes. Histochem. Cell Biol. 141, 17–31 (2014).
Karousou, E. et al. The activity of hyaluronan synthase 2 is regulated by dimerization and ubiquitination. J. Biol. Chem. 285, 23647–23654 (2010).
Oikari, S. et al. UDP-sugar accumulation drives hyaluronan synthesis in breast cancer. Matrix Biol. 67, 63–74 (2018).
Vigetti, D., Viola, M., Karousou, E., De Luca, G. & Passi, A. Metabolic control of hyaluronan synthases. Matrix Biol. 35, 8–13 (2014).
Vigetti, D. et al. Molecular cloning and characterization of UDP-glucose dehydrogenase from the amphibian Xenopus laevis and its involvement in hyaluronan synthesis. J. Biol. Chem. 281, 8254–8263 (2006).
Baggenstoss, B. A. et al. Hyaluronan synthase control of synthesis rate and hyaluronan product size are independent functions differentially affected by mutations in a conserved tandem B-X7-B motif. Glycobiology 27, 154–164 (2016).
Wang, G. et al. Shear stress regulation of endothelial glycocalyx structure is determined by glucobiosynthesis. Arterioscler. Thromb. Vasc. Biol. 40, 350–364 (2020).
Vigetti, D. et al. Role of UDP-N-acetylglucosamine (GlcNAc) and O-GlcNAcylation of hyaluronan synthase 2 in the control of chondroitin sulfate and hyaluronan synthesis. J. Biol. Chem. 287, 35544–35555 (2012).
Vigetti, D. et al. Natural antisense transcript for hyaluronan synthase 2 (HAS2-AS1) induces transcription of HAS2 via protein O-GlcNAcylation. J. Biol. Chem. 289, 28816–28826 (2014).
Chao, H. & Spicer, A. P. Natural antisense mRNAs to hyaluronan synthase 2 inhibit hyaluronan biosynthesis and cell proliferation. J. Biol. Chem. 280, 27513–27522 (2005).
Vigetti, D. et al. Hyaluronan synthesis is inhibited by adenosine monophosphate-activated protein kinase through the regulation of HAS2 activity in human aortic smooth muscle cells. J. Biol. Chem. 286, 7917–7924 (2011).
Stern, R., Kogan, G., Jedrzejas, M. J. & Soltes, L. The many ways to cleave hyaluronan. Biotechnol. Adv. 25, 537–557 (2007).
Stern, R. Devising a pathway for hyaluronan catabolism: are we there yet? Glycobiology 13, 105R–115R (2003).
Bourguignon, V. & Flamion, B. Respective roles of hyaluronidases 1 and 2 in endogenous hyaluronan turnover. FASEB J. 30, 2108–2114 (2016).
Csoka, A. B., Frost, G. I. & Stern, R. The six hyaluronidase-like genes in the human and mouse genomes. Matrix Biol. 20, 499–508 (2001).
Tobisawa, Y. et al. The cell surface hyaluronidase TMEM2 is essential for systemic hyaluronan catabolism and turnover. J. Biol. Chem. 297, 101281 (2021).
Yoshida, H. et al. KIAA1199, a deafness gene of unknown function, is a new hyaluronan binding protein involved in hyaluronan depolymerization. Proc. Natl Acad. Sci. USA 110, 5612–5617 (2013).
Zhang, H. et al. The extracellular matrix integrates mitochondrial homeostasis. Cell 187, 4289–4304.e26 (2024).
Tavianatou, A. G. et al. Hyaluronan: molecular size-dependent signaling and biological functions in inflammation and cancer. FEBS J. 286, 2883–2908 (2019).
Monslow, J., Govindaraju, P. & Pure, E. Hyaluronan — a functional and structural sweet spot in the tissue microenvironment. Front. Immunol. 6, 231 (2015).
Vigetti, D. & Passi, A. Hyaluronan synthases posttranslational regulation in cancer. Adv. Cancer Res. 123, 95–119 (2014).
Berdiaki, A. et al. Hyaluronan and reactive oxygen species signaling-novel cues from the matrix? Antioxidants 12, 824 (2023).
Greyner, H. J., Wiraszka, T., Zhang, L. S., Petroll, W. M. & Mummert, M. E. Inducible macropinocytosis of hyaluronan in B16-F10 melanoma cells. Matrix Biol. 29, 503–510 (2010).
Fraser, J. R., Laurent, T. C. & Laurent, U. B. Hyaluronan: its nature, distribution, functions and turnover. J. Intern. Med. 242, 27–33 (1997).
Screaton, G. R. et al. Genomic structure of DNA encoding the lymphocyte homing receptor CD44 reveals at least 12 alternatively spliced exons. Proc. Natl Acad. Sci. USA 89, 12160–12164 (1992).
Goldstein, L. A. et al. A human lymphocyte homing receptor, the Hermes antigen, is related to cartilage proteoglycan core and link proteins. Cell 56, 1063–1072 (1989).
Aruffo, A., Stamenkovic, I., Melnick, M., Underhill, C. B. & Seed, B. CD44 is the principal cell surface receptor for hyaluronate. Cell 61, 1303–1313 (1990).
Idzerda, R. L. et al. Isolation and DNA sequence of a cDNA clone encoding a lymphocyte adhesion receptor for high endothelium. Proc. Natl Acad. Sci. USA 86, 4659–4663 (1989).
Mackay, C. R. et al. Expression and modulation of CD44 variant isoforms in humans. J. Cell Biol. 124, 71–82 (1994).
Naor, D., Wallach-Dayan, S. B., Zahalka, M. A. & Sionov, R. V. Involvement of CD44, a molecule with a thousand faces, in cancer dissemination. Semin. Cancer Biol. 18, 260–267 (2008).
Zoller, M. CD44, hyaluronan, the hematopoietic stem cell, and leukemia-initiating cells. Front. Immunol. 6, 235 (2015).
Lesley, J., Hyman, R. & Kincade, P. W. CD44 and its interaction with extracellular matrix. Adv. Immunol. 54, 271–335 (1993).
Toyama-Sorimachi, N. & Miyasaka, M. A novel ligand for CD44 is sulfated proteoglycan. Int. Immunol. 6, 655–660 (1994).
Carvalho, A. M., Reis, R. L. & Pashkuleva, I. Hyaluronan receptors as mediators and modulators of the tumor microenvironment. Adv. Healthc. Mater. 12, e2202118 (2023).
Schommer, N. N., Muto, J., Nizet, V. & Gallo, R. L. Hyaluronan breakdown contributes to immune defense against group A Streptococcus. J. Biol. Chem. 289, 26914–26921 (2014).
Scuruchi, M. et al. 6-Mer hyaluronan oligosaccharides modulate neuroinflammation and alpha-synuclein expression in neuron-like SH-SY5Y cells. J. Cell. Biochem. 117, 2835–2843 (2016).
Campo, G. M. et al. 4-mer hyaluronan oligosaccharides stimulate inflammation response in synovial fibroblasts in part via TAK-1 and in part via p38-MAPK. Curr. Med. Chem. 20, 1162–1172 (2013).
Toole, B. P. Hyaluronan and its binding proteins, the hyaladherins. Curr. Opin. Cell Biol. 2, 839–844 (1990).
Albtoush, N. et al. TSG6 hyaluronan matrix remodeling dampens the inflammatory response during colitis. Matrix Biol. 121, 149–166 (2023).
Lee, T. H., Wisniewski, H. G. & Vilcek, J. A novel secretory tumor necrosis factor-inducible protein (TSG-6) is a member of the family of hyaluronate binding proteins, closely related to the adhesion receptor CD44. J. Cell Biol. 116, 545–557 (1992).
Baranova, N. S. et al. The inflammation-associated protein TSG-6 cross-links hyaluronan via hyaluronan-induced TSG-6 oligomers. J. Biol. Chem. 286, 25675–25686 (2011).
Zhang, S., He, H., Day, A. J. & Tseng, S. C. Constitutive expression of inter-α-inhibitor (IαI) family proteins and tumor necrosis factor-stimulated gene-6 (TSG-6) by human amniotic membrane epithelial and stromal cells supporting formation of the heavy chain-hyaluronan (HC-HA) complex. J. Biol. Chem. 287, 12433–12444 (2012).
Zhuo, L. et al. SHAP potentiates the CD44-mediated leukocyte adhesion to the hyaluronan substratum. J. Biol. Chem. 281, 20303–20314 (2006).
Lesley, J. et al. TSG-6 modulates the interaction between hyaluronan and cell surface CD44. J. Biol. Chem. 279, 25745–25754 (2004).
Qadri, M., Almadani, S., Jay, G. D. & Elsaid, K. A. Role of CD44 in regulating TLR2 activation of human macrophages and downstream expression of proinflammatory cytokines. J. Immunol. 200, 758–767 (2018).
Baranova, N. S. et al. Incorporation of pentraxin 3 into hyaluronan matrices is tightly regulated and promotes matrix cross-linking. J. Biol. Chem. 289, 30481–30498 (2014).
Zlibut, A., Bocsan, I. C. & Agoston-Coldea, L. Pentraxin-3 and endothelial dysfunction. Adv. Clin. Chem. 91, 163–179 (2019).
Speeckaert, M. M., Speeckaert, R., Carrero, J. J., Vanholder, R. & Delanghe, J. R. Biology of human pentraxin 3 (PTX3) in acute and chronic kidney disease. J. Clin. Immunol. 33, 881–890 (2013).
Lech, M. et al. Endogenous and exogenous pentraxin-3 limits postischemic acute and chronic kidney injury. Kidney Int. 83, 647–661 (2013).
Hasegawa, K. et al. Versican, a major hyaluronan-binding component in the dermis, loses its hyaluronan-binding ability in solar elastosis. J. Investig. Dermatol. 127, 1657–1663 (2007).
Sin, Y. J. A. et al. Noncovalent hyaluronan crosslinking by TSG-6: modulation by heparin, heparan sulfate, and PRG4. Front. Mol. Biosci. 9, 990861 (2022).
Chanmee, T., Ontong, P., Kimata, K. & Itano, N. Key roles of hyaluronan and its CD44 receptor in the stemness and survival of cancer stem cells. Front. Oncol. 5, 180 (2015).
Liu, H. et al. Oct4 regulates the miR-302 cluster in P19 mouse embryonic carcinoma cells. Mol. Biol. Rep. 38, 2155–2160 (2011).
Moretto, P. et al. Regulation of hyaluronan synthesis in vascular diseases and diabetes. J. Diabetes Res. 2015, 1–9 (2015).
Lagendijk, A. K., Szabo, A., Merks, R. M. & Bakkers, J. Hyaluronan: a critical regulator of endothelial-to-mesenchymal transition during cardiac valve formation. Trends Cardiovasc. Med. 23, 135–142 (2013).
Lagendijk, A. K., Goumans, M. J., Burkhard, S. B. & Bakkers, J. MicroRNA-23 restricts cardiac valve formation by inhibiting Has2 and extracellular hyaluronic acid production. Circ. Res. 109, 649–657 (2011).
Bakkers, J. et al. Has2 is required upstream of Rac1 to govern dorsal migration of lateral cells during zebrafish gastrulation. Development 131, 525–537 (2004).
Nandadasa, S. et al. The versican-hyaluronan complex provides an essential extracellular matrix niche for Flk1+ hematoendothelial progenitors. Matrix Biol. 97, 40–57 (2021).
Sherpa, A. D. et al. Integrity of white matter is compromised in mice with hyaluronan deficiency. Neurochem. Res. 45, 53–67 (2020).
Sanketi, B. D. et al. Pitx2 patterns an accelerator-brake mechanical feedback through latent TGFβ to rotate the gut. Science 377, eabl3921 (2022).
Sivakumar, A. et al. Midgut laterality is driven by hyaluronan on the right. Dev. Cell 46, 533–551.e5 (2018).
Munjal, A., Hannezo, E., Tsai, T. Y., Mitchison, T. J. & Megason, S. G. Extracellular hyaluronate pressure shaped by cellular tethers drives tissue morphogenesis. Cell 184, 6313–6325 e6318 (2021).
Pardue, E. L., Ibrahim, S. & Ramamurthi, A. Role of hyaluronan in angiogenesis and its utility to angiogenic tissue engineering. Organogenesis 4, 203–214 (2008).
Vernon, R. B. et al. Autocrine hyaluronan influences sprouting and lumen formation during HUVEC tubulogenesis in vitro. J. Histochem. Cytochem. 69, 415–428 (2021).
Lokeshwar, V. B. & Selzer, M. G. Differences in hyaluronic acid-mediated functions and signaling in arterial, microvessel, and vein-derived human endothelial cells. J. Biol. Chem. 275, 27641–27649 (2000).
Queisser, K. A., Mellema, R. A. & Petrey, A. C. Hyaluronan and its receptors as regulatory molecules of the endothelial interface. J. Histochem. Cytochem. 69, 25–34 (2021).
Parnigoni, A. et al. Hyaluronan in pathophysiology of vascular diseases: specific roles in smooth muscle cells, endothelial cells, and macrophages. Am. J. Physiol. Cell Physiol. 323, C505–C519 (2022).
Murphy, J. F. et al. Engagement of CD44 modulates cyclooxygenase induction, VEGF generation, and proliferation in human vascular endothelial cells. FASEB J. 19, 446–448 (2005).
Rodgers, L. S. et al. Depolymerized hyaluronan induces vascular endothelial growth factor, a negative regulator of developmental epithelial-to-mesenchymal transformation. Circ. Res. 99, 583–589 (2006).
Little, M. H. & McMahon, A. P. Mammalian kidney development: principles, progress, and projections. Cold Spring Harb. Perspect. Biol. 4, a008300 (2012).
McMahon, A. P. Development of the mammalian kidney. Curr. Top. Dev. Biol. 117, 31–64 (2016).
Li, H., Hohenstein, P. & Kuure, S. Embryonic kidney development, stem cells and the origin of Wilms tumor. Genes 12, 318 (2021).
Pichel, J. G. et al. Defects in enteric innervation and kidney development in mice lacking GDNF. Nature 382, 73–76 (1996).
Saxen, L. & Sariola, H. Early organogenesis of the kidney. Pediatr. Nephrol. 1, 385–392 (1987).
Linton, J. M., Martin, G. R. & Reichardt, L. F. The ECM protein nephronectin promotes kidney development via integrin α8β1-mediated stimulation of Gdnf expression. Development 134, 2501–2509 (2007).
Motamedi, F. J. et al. WT1 controls antagonistic FGF and BMP-pSMAD pathways in early renal progenitors. Nat. Commun. 5, 4444 (2014).
Kanwar, Y. S. et al. Role of extracellular matrix, growth factors and proto-oncogenes in metanephric development. Kidney Int. 52, 589–606 (1997).
Hochane, M. et al. Single-cell transcriptomics reveals gene expression dynamics of human fetal kidney development. PLoS Biol. 17, e3000152 (2019).
Wang, G. et al. Spatial dynamic metabolomics identifies metabolic cell fate trajectories in human kidney differentiation. Cell Stem Cell 29, 1580–1593 e1587 (2022).
Soulie, P., Chassot, A., Ernandez, T., Montesano, R. & Feraille, E. Spatially restricted hyaluronan production by Has2 drives epithelial tubulogenesis in vitro. Am. J. Physiol. Cell Physiol. 307, C745–C759 (2014).
Belsky, E. & Toole, B. P. Hyaluronate and hyaluronidase in the developing chick embryo kidney. Cell Differ. 12, 61–66 (1983).
Grobstein, C. Inductive interaction in the development of the mouse metanephros. J. Exp. Zool. 130, 319–339 (1955).
Kobayashi, A. et al. Identification of a multipotent self-renewing stromal progenitor population during mammalian kidney organogenesis. Stem Cell Rep. 3, 650–662 (2014).
Kobayashi, A. et al. Six2 defines and regulates a multipotent self-renewing nephron progenitor population throughout mammalian kidney development. Cell Stem Cell 3, 169–181 (2008).
Shakya, R. et al. The role of GDNF in patterning the excretory system. Dev. Biol. 283, 70–84 (2005).
Short, K. M. et al. Global quantification of tissue dynamics in the developing mouse kidney. Dev. Cell 29, 188–202 (2014).
Rosines, E., Schmidt, H. J. & Nigam, S. K. The effect of hyaluronic acid size and concentration on branching morphogenesis and tubule differentiation in developing kidney culture systems: potential applications to engineering of renal tissues. Biomaterials 28, 4806–4817 (2007).
Pohl, M., Sakurai, H., Stuart, R. O. & Nigam, S. K. Role of hyaluronan and CD44 in in vitro branching morphogenesis of ureteric bud cells. Dev. Cell 224, 312–325 (2000).
Toole, B. P. Hyaluronan in morphogenesis. J. Intern. Med. 242, 35–40 (1997).
Faa, G. et al. The role of immunohistochemistry in the study of the newborn kidney. J. Matern. Fetal Neonatal Med. 25, 135–138 (2012).
Rozario, T. & DeSimone, D. W. The extracellular matrix in development and morphogenesis: a dynamic view. Dev. Cell 341, 126–140 (2010).
Knepper, M. A., Saidel, G. M., Hascall, V. C. & Dwyer, T. Concentration of solutes in the renal inner medulla: interstitial hyaluronan as a mechano-osmotic transducer. Am. J. Physiol. Renal Physiol. 284, F433–F446 (2003).
Wells, A. F. et al. The localization of hyaluronan in normal and rejected human kidneys. Transplantation 50, 240–243 (1990).
Goransson, V., Johnsson, C., Nylander, O. & Hansell, P. Renomedullary and intestinal hyaluronan content during body water excess: a study in rats and gerbils. J. Physiol. 542, 315–322 (2002).
Rugheimer, L. et al. Hyaluronan synthases and hyaluronidases in the kidney during changes in hydration status. Matrix Biol. 28, 390–395 (2009).
Stridh, S., Palm, F. & Hansell, P. Renal interstitial hyaluronan: functional aspects during normal and pathological conditions. Am. J. Physiol. Regul. Integr. Comp. Physiol. 302, R1235–R1249 (2012).
Hansell, P., Goransson, V., Odlind, C., Gerdin, B. & Hallgren, R. Hyaluronan content in the kidney in different states of body hydration. Kidney Int. 58, 2061–2068 (2000).
Goransson, V. et al. Renomedullary interstitial cells in culture; the osmolality and oxygen tension influence the extracellular amounts of hyaluronan and cellular expression of CD44. Matrix Biol. 20, 129–136 (2001).
Johnsson, C., Tufveson, G., Wahlberg, J. & Hallgren, R. Experimentally-induced warm renal ischemia induces cortical accumulation of hyaluronan in the kidney. Kidney Int. 50, 1224–1229 (1996).
van den Berg, B. M. et al. Glomerular function and structural integrity depend on hyaluronan synthesis by glomerular endothelium. J. Am. Soc. Nephrol. 30, 1886–1897 (2019).
Rabelink, T. J. et al. Heparanase: roles in cell survival, extracellular matrix remodelling and the development of kidney disease. Nat. Rev. Nephrol. 13, 201–212 (2017).
Dane, M. J. et al. Glomerular endothelial surface layer acts as a barrier against albumin filtration. Am. J. Pathol. 182, 1532–1540 (2013).
Dane, M. J. et al. A microscopic view on the renal endothelial glycocalyx. Am. J. Physiol. Renal Physiol. 308, F956–F966 (2015).
Ramiro-Diaz, J. et al. Luminal endothelial lectins with affinity for N-acetylglucosamine determine flow-induced cardiac and vascular paracrine-dependent responses. Am. J. Physiol. 299, H743–H751 (2010).
Parthasarathy, N. & Spiro, R. G. Characterization of the glycosaminoglycan component of the renal glomerular basement membrane and its relationship to the peptide portion. J. Biol. Chem. 256, 507–513 (1981).
Smith, D. W., Azadi, A., Lee, C. J. & Gardiner, B. S. Spatial composition and turnover of the main molecules in the adult glomerular basement membrane. Tissue Barriers 11, 2110798 (2023).
Balaji, S. et al. Interleukin-10-mediated regenerative postnatal tissue repair is dependent on regulation of hyaluronan metabolism via fibroblast-specific STAT3 signaling. FASEB J. 31, 868–881 (2017).
Wang, X. et al. High-molecular weight hyaluronan attenuates tubulointerstitial scarring in kidney injury. JCI Insight 5, e136345 (2020).
Zhao, Y. B., Wei, W., Lin, X. X., Chai, Y. F. & Jin, H. The role of histone H3 methylation in acute kidney injury. Drug. Des. Devel. Ther. 16, 2453–2461 (2022).
Yu, C. et al. Histone demethylase JMJD3 protects against renal fibrosis by suppressing TGFβ and Notch signaling and preserving PTEN expression. Theranostics 11, 2706–2721 (2021).
Nakka, K. et al. JMJD3 activated hyaluronan synthesis drives muscle regeneration in an inflammatory environment. Science 377, 666–669 (2022).
Kato, T. et al. Adipose tissue-derived stem cells suppress acute cellular rejection by TSG-6 and CD44 interaction in rat kidney transplantation. Transplantation 98, 277–284 (2014).
Dyer, D. P. et al. TSG-6 inhibits neutrophil migration via direct interaction with the chemokine CXCL8. J. Immunol. 192, 2177–2185 (2014).
Salman, L. et al. Hyaluronan inhibition as a therapeutic target for diabetic kidney disease: what is next? Kidney360 4, e851–e860 (2023).
Selman, G. et al. A hyaluronan synthesis inhibitor delays the progression of diabetic kidney disease in a mouse experimental model. Kidney360 2, 809–818 (2021).
Khramova, A. et al. Proteoglycans contribute to the functional integrity of the glomerular endothelial cell surface layer and are regulated in diabetic kidney disease. Sci. Rep. 11, 8487 (2021).
Morimoto, K. et al. Renal arteriolar hyalinosis, not intimal thickening in large arteries, is associated with cardiovascular events in people with biopsy-proven diabetic nephropathy. Diabet. Med. 37, 2143–2152 (2020).
Oguchi, H. et al. Vasa recta hyalinosis reflects severe arteriolopathy in renal allografts. Clin. Exp. Nephrol. 23, 799–806 (2019).
Rouschop, K. M. et al. Protection against renal ischemia reperfusion injury by CD44 disruption. J. Am. Soc. Nephrol. 16, 2034–2043 (2005).
Rosser, J. I. et al. Oral hymecromone decreases hyaluronan in human study participants. J. Clin. Invest. 132, e157983 (2022).
Dogne, S. et al. Hyaluronidase 1 deficiency preserves endothelial function and glycocalyx integrity in early streptozotocin-induced diabetes. Diabetes 65, 2742–2753 (2016).
Martin, D. C. et al. A mouse model of human mucopolysaccharidosis IX exhibits osteoarthritis. Hum. Mol. Genet. 17, 1904–1915 (2008).
Natowicz, M. R. et al. Clinical and biochemical manifestations of hyaluronidase deficiency. N. Engl. J. Med. 335, 1029–1033 (1996).
Onclinx, C. et al. Deficiency in mouse hyaluronidase 2: a new mechanism of chronic thrombotic microangiopathy. Haematologica 100, 1023–1030 (2015).
Jin, C. & Zong, Y. The role of hyaluronan in renal cell carcinoma. Front. Immunol. 14, 1127828 (2023).
Wang, J. et al. Targeting hyaluronic acid synthase-3 (HAS3) for the treatment of advanced renal cell carcinoma. Cancer Cell Int. 22, 421 (2022).
Jokelainen, O. et al. Cellular hyaluronan is associated with a poor prognosis in renal cell carcinoma. Urol. Oncol. 38, 686 e611–686 e622 (2020).
Chi, A. et al. Molecular characterization of kidney cancer: association of hyaluronic acid family with histological subtypes and metastasis. Cancer 118, 2394–2402 (2012).
Moran, H. et al. Glycocalyx mechanotransduction mechanisms are involved in renal cancer metastasis. Matrix Biol. 13, 100100 (2022).
Sun, M. et al. MicroRNA-125a suppresses cell migration, invasion, and regulates hyaluronic acid synthase 1 expression by targeting signal transducers and activators of transcription 3 in renal cell carcinoma cells. J. Cell. Biochem. 120, 1894–1902 (2019).
Chanmee, T., Ontong, P. & Itano, N. Hyaluronan: a modulator of the tumor microenvironment. Cancer Lett. 375, 20–30 (2016).
Jin, J. et al. Sunitinib resistance in renal cell carcinoma: from molecular mechanisms to predictive biomarkers. Drug Resist. Updat. 67, 100929 (2023).
Rini, B. I. & Atkins, M. B. Resistance to targeted therapy in renal-cell carcinoma. Lancet Oncol. 10, 992–1000 (2009).
Lasorsa, F. et al. Cancer stem cells in renal cell carcinoma: origins and biomarkers. Int. J. Mol. Sci. 24, 13179 (2023).
Mikami, S. et al. Expression of TNF-α and CD44 is implicated in poor prognosis, cancer cell invasion, metastasis and resistance to the sunitinib treatment in clear cell renal cell carcinomas. Int. J. Cancer 136, 1504–1514 (2015).
Lokeshwar, V. B., Mirza, S. & Jordan, A. Targeting hyaluronic acid family for cancer chemoprevention and therapy. Adv. Cancer Res. 123, 35–65 (2014).
Morosi, L. et al. PEGylated recombinant human hyaluronidase (PEGPH20) pre-treatment improves intra-tumour distribution and efficacy of paclitaxel in preclinical models. J. Exp. Clin. Cancer Res. 40, 286 (2021).
Kim, D. K. et al. Scalable production of a multifunctional protein (TSG-6) that aggregates with itself and the CHO cells that synthesize it. PLoS ONE 11, e0147553 (2016).
Moriya, T. et al. Arteriolar hyalinosis predicts increase in albuminuria and GFR decline in normo- and microalbuminuric Japanese patients with type 2 diabetes. Diabetes Care 40, 1373–1378 (2017).
Mencke, R. et al. Klotho deficiency induces arteriolar hyalinosis in a trade-off with vascular calcification. Am. J. Pathol. 189, 2503–2515 (2019).
Menon, R. et al. Defining the molecular correlate of arteriolar hyalinosis in kidney disease progression by integration of single cell transcriptomic analysis and pathology scoring. Preprint at medRxiv https://doi.org/10.1101/2023.06.14.23291150 (2023).
Hall, J. E., do Carmo, J. M., da Silva, A. A., Wang, Z. & Hall, M. E. Obesity, kidney dysfunction and hypertension: mechanistic links. Nat. Rev. Nephrol. 15, 367–385 (2019).
Dwyer, T. M. et al. Distribution of renal medullary hyaluronan in lean and obese rabbits. Kidney Int. 58, 721–729 (2000).
Alonsogalicia, M., Dwyer, T. M., Herrera, G. A. & Hall, J. E. Increased hyaluronic-acid in the inner renal medulla of obese dogs. Hypertension 25, 888–892 (1995).
Hansell, P. et al. Renomedullary interstitial cells regulate hyaluronan turnover depending on growth media osmolality suggesting a role in renal water handling. Acta Physiol. Scand. 165, 115–116 (1999).
Dwyer, T. M., Stec, D. E., Hall, J. E. & Csongradi, E. Hyaluronan is required for the renal medulla to maximally concentrate urine. FASEB J. https://doi.org/10.1096/fasebj.25.1_supplement.1079.5 (2011).
Adams, J. D. et al. Comparing thirst and spot urine concentrations in humans of differing body sizes: an observational study. Physiol. Behav. 245, 113673 (2022).
Acknowledgements
The authors thank S. M. Chuva de Sousa Lopes for providing the fetal tissue sample used in Fig. 4; W. M. P. J. Sol for processing and immune fluorescence staining of the tissue sample, and M. Zuurmond for editing and drafting the originally submitted versions of the figures. The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) is supported by Novo Nordisk Foundation grants (NNF21CC0073729). Some of the authors’ work was funded through the Dutch Kidney Foundation (GLYCOREN Consortium grant no. CP09.03).
Author information
Authors and Affiliations
Contributions
T.J.R. and B.M.v.d.B. primarily researched the data for the article and wrote the text. G.W. and J.v.d.V. contributed to discussions on the interpretation of the literature and reviewed and edited the manuscript before submission.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Nephrology thanks Michael Goligorsky, Peter Hansell and Thomas Wight for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Rabelink, T.J., Wang, G., van der Vlag, J. et al. The roles of hyaluronan in kidney development, physiology and disease. Nat Rev Nephrol (2024). https://doi.org/10.1038/s41581-024-00883-5
Accepted:
Published:
DOI: https://doi.org/10.1038/s41581-024-00883-5