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 -- 2299 2023-01-17 03:42:55 |
2 update references and layout Meta information modification 2299 2023-01-17 04:06:20 |

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.
Hua, S.H.;  Viera, M.;  Yip, G.W.;  Bay, B.H. Glycosaminoglycans in Metastatic Renal Cell Carcinoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/40258 (accessed on 07 July 2024).
Hua SH,  Viera M,  Yip GW,  Bay BH. Glycosaminoglycans in Metastatic Renal Cell Carcinoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/40258. Accessed July 07, 2024.
Hua, San Hue, Maximillian Viera, George W. Yip, Boon Huat Bay. "Glycosaminoglycans in Metastatic Renal Cell Carcinoma" Encyclopedia, https://encyclopedia.pub/entry/40258 (accessed July 07, 2024).
Hua, S.H.,  Viera, M.,  Yip, G.W., & Bay, B.H. (2023, January 17). Glycosaminoglycans in Metastatic Renal Cell Carcinoma. In Encyclopedia. https://encyclopedia.pub/entry/40258
Hua, San Hue, et al. "Glycosaminoglycans in Metastatic Renal Cell Carcinoma." Encyclopedia. Web. 17 January, 2023.
Glycosaminoglycans in Metastatic Renal Cell Carcinoma
Edit

Renal cell carcinoma (RCC) makes up the majority of kidney cancers, with a poor prognosis for metastatic RCC (mRCC). Challenges faced in the management of mRCC, include a lack of reliable prognostic markers and biomarkers for precise monitoring of disease treatment, together with the potential risk of toxicity associated with more recent therapeutic options. Glycosaminoglycans (GAGs) are a class of carbohydrates that can be categorized into four main subclasses, viz., chondroitin sulfate, hyaluronic acid, heparan sulfate and keratan sulfate. GAGs are known to be closely associated with cancer progression and modulation of metastasis by modification of the tumor microenvironment. Alterations of expression, composition and spatiotemporal distribution of GAGs in the extracellular matrix (ECM), dysregulate ECM functions and drive cancer invasion.

glycosaminoglycans renal cell carcinoma metastasis

1. Metastatic Renal Cell Carcinoma

Renal cell carcinoma (RCC) comprises more than 90% of cases of kidney cancer, with clear cell RCC (ccRCC) being the most common type of RCC and making up the majority of cancer-related deaths [1][2]. The “founding event” of ccRCC is often attributed to a mutation in the von Hippel-Lindau (VHL) tumor suppressor gene [3], although by itself is insufficient to cause ccRCC. The prognosis for RCC is poor, especially for metastatic RCC (mRCC). The overall 5-year survival rate for RCC patients is 74% and decreases to only 8% for patients with mRCC [4][5]. Despite improvements in early detection techniques and considerable progress in systemic treatment, a quarter of patients with localized RCC still develops metastatic deposits at distant sites following surgical removal of the primary tumor (post-nephrectomy) [6][7]. Distant metastases are mostly observed in the lymph nodes, lungs, liver, bone and brain [8].
For early or resectable RCC, nephrectomy is usually performed in the management of this cancer. As RCC is usually resistant to conventional chemotherapy and radiotherapy, the standard treatments for mRCC have been interleukin-2 and interferon cytokine-based therapies, until the availability of targeted therapies [2][9]. Several targeted treatments are available for mRCC management, the most common being tyrosine kinase inhibitors targeting Vascular endothelial growth factor (VEGF) signaling, such as sunitinib [10][11] and sorafenib [12]. Sunitinib is commonly used as a first line treatment option for RCC, with a higher response rate and longer progression-free survival than the conventionally used interferon α as observed during a Phase 3 clinical trial [10]. Moreover, poor-risk RCC patients were also observed to show responses to the drug in the same study. Although sunitinib is still used in VEGFR-targeted therapy for advanced RCC [13], newer generation of multiple VEGF kinase inhibitors, such as Lenvatinib, has been found to be more effective [14].
Another drug target is the mammalian target of rapamycin (mTOR) pathway, which regulates cell proliferation and tumor metabolism [15]. Temsirolimus, a specific inhibitor of mTOR, has been used as both first line and second line treatment options in advanced RCC [16][17]. This intravenous drug has also been shown to achieve prolonged survival over interferon α among mRCC patients in a Phase 3 clinical trial [18][19]. However, despite the clinically beneficial outcomes that these targeted treatments offer, nearly all RCC patients develop resistance to both VEGF-targeted and mTOR-targeted therapies. Thus, a combination of VEGF and mTOR inhibitors has been administered as a strategy to delay drug resistance to either class of the inhibitors [20][21].
Another treatment modality employed is immunotherapy targeting the programmed cell death protein 1 (PD1) and its ligand PDL1, as demonstrated by the effects of the drug nivolumab [22]. PDL1 is overexpressed in cancer cells, and inhibiting PD1-PDL1 interaction promotes T-cell activation and killing of cancer cells. Nivolumab, used as second line treatment for mRCC, has been reported to offer a longer overall survival and higher response rates, with fewer adverse effects and a better quality of life, compared to the mTOR inhibitor Everolimus [23]. A meta-analysis comprising 5121 patients with mRCC from six clinical trials, revealed that Nivolumab plus cabozantinib (an oral inhibitor of multiple tyrosine kinases) was associated with the highest likelihood of patients having maximal overall survival, while the combination of Lenvatinib plus Pembrolizumab (a humanized antibody used in cancer immunotherapy), the highest likelihood of progression free survival [24]. In fact, the European Association of Urology Guidelines has recommended combination therapies of Axitinib plus Pembrolizumab, Cabozantinib plus Nivolumab, and Lenvatinib plus Pembrolizumab for advanced RCC [14]. Another combination therapy which has undergone a phase III clinical trial include, randomization of 873 patients who received either axitinib and avelumab or sunitinib [25]. A recent report has also shown evidence for clinically meaningful and durable benefits in advanced RCC patients treated with Nivolumab plus Ipilimumab (a monoclonal antibody that targets CTLA-4) [26].

2. Glycosaminoglycans

Glycosaminoglycans (GAGS) are linear polysaccharides that consist of repeating disaccharide units of uronic acid and an amino sugar [27]. They are found in almost every mammalian tissue, providing structural scaffolding and hydration to the cells [28]. Figure 1 is an illustration of the four main classes of glycosaminoglycans: hyaluronic acid (HA), chondroitin sulfate (CS), heparan sulfate (HS), and keratan sulfate (KS). Monomers of the disaccharide building blocks consist of GlcA (d-glucuronic acid) and GlcNAc (N-acetyl-d-glucosamine) for HA; GlcA and GalNAc (N-acetyl-d-galactosamine) for CS; GlcA or IdoA (l-iduronic acid) and GlcNAc or GlcN (d-glucosamine) for HS; Gal (d-galactose) and GlcNAc for KS. GAGs are highly polar and negatively charged with the polysaccharide lengths generally varying between 4 and 200 mer [29]. With the exception of HA, GAGs contain sulfate groups attached at specific sites [30]. The sulfate groups are added onto the GAGs chain through post-polymerization modifications [31]. O-sulfotransferases mediate the sulfation of CS and KS while the sulfation of HS is controlled by N-sulfotransferases, C5 epimerases as well as O-sulfotransferases [29][32].
Figure 1. Structure of the four main glycosaminoglycans (GAGs). Disaccharide monomers of Hyaluronic acid (A); Chondroitin sulfate (B); Heparan sulfate (C) and Keratan sulfate (D). The possible sulfate sites are denoted with Ri, the superscript ‘i’ indicate the Carbon position where the sulfate group is esterified; R = H or SO3H. Representative Glycosaminoglycans polysaccharides of HA, CS, HS and KS consist of repeating disaccharides monomers with various sulfation patterns (E).
HA (the only GAG known not to have any sulfation sites) has a crucial role in cushioning and lubricating the body that is attributable to its highly hydrophilic property, and is therefore found in abundance in the eyes, joints, and heart valves [33]. HA is also abundant in the skin and important in wound healing [34]. KS is present in the cornea, cartilage, and bones, and associated with disorders such as macular corneal dystrophy and osteoarthritis [35]. HS is usually located in the extracellular matrix (ECM), and is highly involved in tumorigenesis [36][37][38]. CS is an important structural component of cartilage, providing much of their resistance to compression [39]. CS can interact with various biomolecules and form proteoglycans (PGs) with proteins, the major components of the extracellular matrix and drive crucial biological activities. The various sulfation patterns of CS could code for different biological regulatory functions [40][41]. For instance, C4S is known to be important for cartilage regeneration and observed to be downregulated in degraded osteoarthritic cartilage [42]. C4,6S augmented cartilage generation through enhancing type II collagen production [43]. C4,6S, but not C4S or C6S, was reported to be able to interact with several neurotrophic factors to stimulate neurite outgrowth [40].
Interestingly, it is well established that GAGs are involved in cancer cell growth, signalling, and metastasis [28][44]. HA levels are significantly elevated in breast [45], lung [46], and ovarian cancers [47]. Certain sulfation motifs of exogenous CS were shown to induce apoptosis and inhibit the growth of triple negative breast cancer cells [48]. Dermatan sulfate was observed to be elevated with changes in the sulfation profiles in the stroma of certain cancers such as liver [49], lung [50][51], pancreatic [52], colorectal [53] and gastric [54] cancers.

3. GAGs and Metastasis

Cancer is a complex disease where malignant cells could acquire the ability to metastasize to distant sites, thus accounting for the majority of cancer-related morbidity and mortality. The fundamental processes of migration and invasion are crucial for cancer metastasis, which is usually the primary cause of death in cancer patients [55]. The ECM, which is composed mainly of GAGs and their PGs, would therefore play a significant role in controlling cell behavior and movement [56]. GAGs are endowed with rigidity property, thus providing structural integrity to the cells and passageways in the ECM between cells [30]. Dysregulation of ECM modelling occurs during tumorigenesis and metastasis, leading to changes in the tumor microenvironment (TME) and loss of tissue homeostasis [57][58]. Disorganization in the ECM GAGs/PGs expression, composition and spatiotemporal distribution are the main causes of the dysregulation of ECM functions and the driver for cancer invasion [59]. GAGs modulate cancer invasion through binding with various growth factors, adhesion molecules and cytokines [60].
HA is the only GAG that does not form PGs with any protein through covalent bonding, and therefore not sulfated at all [31]. Increased HA synthesis through overexpression of Hyaluronan Synthases (HS) was observed to promote cancer growth and metastasis in xenograft models of breast, prostate, and colon cancers [61][62][63][64]. The presence of HA provides cancer cells with a highly hydrated and malleable ECM which is essential for changes in cell shapes and tissue penetration during invasion [65][66][67]. Pericellular HA surrounding metastatic cancer cells could facilitate adhesion of cancer cells to endothelial cells at the metastatic site [68][69] (Figure 2). Moreover, HA can interact with various cell-surface receptors, notably CD44 and RHAMM, which are well established to be involved in cancer cell survival, motility, and metastasis [70][71]. Studies have shown that disruption of the binding of HA to either CD44 or RHAMM receptors [72][73][74], would suppress the development of metastatic nodules in mice. HA-CD44 interaction has been reported to stimulate Matrix Metalloproteinase 2 (MMP2) and MMP9 expression and their cell-surface presentation [75]. These MMPs play important roles in cancer invasion, as they aid in digesting through the ECM barrier (which is essential in preventing cancer cells from escaping their primary tissue architecture) and in facilitating the growth of cancer cells at metastatic sites [76][77]. HA bound to RHAMM could induce the activation of FAK which is required for actin filament and microtubule rearrangements as well as cancer cell motility [78][79][80][81].
Figure 2. Proposed mechanisms of the involvement of GAGs in cancer metastasis. In metastatic renal cancer, HA, HS and HS present on the cell surface layer promote cancer cell invasion and metastatic capabilities. (A) During metastasis, cancer cells traveling in the blood vessel adhere to the endothelial cells through the binding and interaction between cell surface CD44 and HA present in the ECM. (B) HA-bound CD44 receptors on cancer cells activates MAPK-ERK1/2 and AKT signaling pathways to promote cell adhesion, migration and invasion. CD44 also increases MMP2 and MMP9 expression and secretion to help digest and remodel ECM proteins at the metastatic site. Cell surface RHAMM, upon the binding of HA in the ECM, activates FAK-Src signaling pathways to help cancer cells migrate through the blood vessel and start colonization at the metastatic site. (C) Cell surface proteoglycan of CS and HS, such as Versican and Syndecan-1, promote cancer invasion though activating Snail, EGFR and AKT signaling pathways.
CS, HS and KS can be sulfated and form PGs with the core proteins through covalent binding at the Serine residues [82]. Alterations in cell surface CS expression, sulfation patterns and consequently, ECM-degradative enzymes, such as MMPs, would result in changes of cell invasiveness and disruption of cell-matrix interactions [27][83]. In vitro studies on breast cancer revealed that a higher CS expression in tumor cells was concomitant with increased cell proliferation, migration and invasion [84][85][86][87]. At the tissue level, CS in general was observed to be significantly elevated in the stromal compartment of breast tumors [88][89]. The sulfation patterns of CS also elicited various effects on cancer invasion [83]. For instance, elevated expression of non-sulfated chondroitin in prostate cancer was associated with adverse clinicopathological parameters [90]. C4,6S was noted to suppress cancer cell invasion through inhibiting Wnt/β- Catenin signaling [91] and enhancing the retention of tissue inhibitor of metalloproteinases (TIMP)-3, which disrupt ECM processing and cell mobility [92]. On the other hand, C4,6S expression could promote ovarian cancer metastasis through interacting with VEGF, HGF [93][94] and P-Selectin [95], which support survival of the circulating cancer cells and tissue colonization. C6S bound to CD44 has been reported to promote cancer cell adhesion and migration [96]. C4S is known to suppress cancer invasiveness and inhibit cathepsin S activity which regulate cell–cell and cell-ECM contacts [97][98]. CS proteoglycans (CSPGs) also participate in cancer cells migration, invasion, and metastasis. A large ECM CSPG (Versican) was observed to promote cancer epithelial-to-mesenchymal transition (EMT) and metastasis through EGFR/AKT [99], Snail/PAPSS2 [100] and TGFβ/NK-κB signaling [101] in liver, breast and ovarian cancers. Intracellular CSPG Serglycin has been reported to interact with CD44 [102][103] and activate IL-8 [104] signaling pathways to enhance cell migration and metastasis in lung and breast cancer.
HS is present at the ECM interface to modulate various types of cell-ECM interactions [105]. The capability of HS to bind to various chemokines, growth factors, morphogens, enzymes and ECM proteins, confer functional properties such as controlling cancer migration, EMT and metastasis [28]. Changes in HS sulfation patterns could also affect cancer cell invasion and metastasis [106][107]. Reduction in 6-O-Sulfation of HS has been observed to augment VEGF and FGF induced cell invasion in RCC [108]. On the other hand, an increase in 3-O-Sulfation could enhance the EMT and invasion capacity of pancreatic cancer cells [109]. Cell surface HS/CS Proteoglycans (HSPGs) Syndecan-1 is known to increase cancer stemness and invasiveness through stimulating the Notch and EGFR signaling pathways and regulation of the focal adhesion kinase-Wnt signaling axis [110][111]. In contrast, ECM HSPG Perlecan has been shown to inhibit cancer cell invasion and digested by MMP7 during FAK driven invasion in prostate cancer [112].
KS expression was reported to be increased in pancreatic tumor tissues compared to normal adjacent tissues and stroma, with KS expression being higher in lung metastatic sites compared to the primary pancreatic tumor [113]. KSPG Lumican has been shown to inhibit lung cancer invasion through binding with p120-catenin, which prevent activation of Rho GTPases, FAK and cytoskeletal re-organization [114]. On the other hand, highly glycosylated ECM Lumincan promote colon cancer cells migration through binding with cell surface integrins, and activating actin cytoskeleton remodeling [115][116].

References

  1. Hsieh, J.J.; Purdue, M.P.; Signoretti, S.; Swanton, C.; Albiges, L.; Schmidinger, M.; Heng, D.Y.; Larkin, J.; Ficarra, V. Renal cell carcinoma. Nat. Rev. Dis. Prim. 2017, 3, 17009.
  2. Rini, B.I.; Campbell, S.C.; Escudier, B. Renal cell carcinoma. Lancet 2009, 373, 1119–1132.
  3. Frew, I.J.; Moch, H. A clearer view of the molecular complexity of clear cell renal cell carcinoma. Annu. Rev. Pathol. 2015, 10, 263–289.
  4. Surveillance, E.; Program, E.R. SEER Stat Fact Sheets: Kidney and Renal Pelvis Cancer; National Cancer Institute: Bethesda, MD, USA, 2017.
  5. Kosary, C. Kidney and renal pelvis. SEER Cancer Statistics Review, 1973–1990; National Cancer Institute: Bethesda, MD, USA, 1993.
  6. Dabestani, S.; Thorstenson, A.; Lindblad, P.; Harmenberg, U.; Ljungberg, B.; Lundstam, S. Renal cell carcinoma recurrences and metastases in primary non-metastatic patients: A population-based study. World J. Urol. 2016, 34, 1081–1086.
  7. Rabinovitch, R.A.; Zelefsky, M.J.; Gaynor, J.J.; Fuks, Z. Patterns of failure following surgical resection of renal cell carcinoma: Implications for adjuvant local and systemic therapy. J. Clin. Oncol. 1994, 12, 206–212.
  8. McKay, R.R.; Kroeger, N.; Xie, W.; Lee, J.L.; Knox, J.J.; Bjarnason, G.A.; MacKenzie, M.J.; Wood, L.; Srinivas, S.; Vaishampayan, U.N.; et al. Impact of bone and liver metastases on patients with renal cell carcinoma treated with targeted therapy. Eur. Urol. 2014, 65, 577–584.
  9. Ljungberg, B.; Cowan, N.C.; Hanbury, D.C.; Hora, M.; Kuczyk, M.A.; Merseburger, A.S.; Patard, J.J.; Mulders, P.F.; Sinescu, I.C. EAU guidelines on renal cell carcinoma: The 2010 update. Eur. Urol. 2010, 58, 398–406.
  10. Motzer, R.J.; Hutson, T.E.; Tomczak, P.; Michaelson, M.D.; Bukowski, R.M.; Rixe, O.; Oudard, S.; Negrier, S.; Szczylik, C.; Kim, S.T.; et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N. Engl. J. Med. 2007, 356, 115–124.
  11. Deng, H.; Liu, W.; He, T.; Hong, Z.; Yi, F.; Wei, Y.; Zhang, W. Comparative Efficacy, Safety, and Costs of Sorafenib vs. Sunitinib as First-Line Therapy for Metastatic Renal Cell Carcinoma: A Systematic Review and Meta-Analysis. Front. Oncol. 2019, 9, 479.
  12. Escudier, B.; Eisen, T.; Stadler, W.M.; Szczylik, C.; Oudard, S.; Siebels, M.; Negrier, S.; Chevreau, C.; Solska, E.; Desai, A.A.; et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N. Engl. J. Med. 2007, 356, 125–134.
  13. Choueiri, T.K.; Escudier, B.; Powles, T.; Mainwaring, P.N.; Rini, B.I.; Donskov, F.; Hammers, H.; Hutson, T.E.; Lee, J.L.; Peltola, K.; et al. Cabozantinib versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2015, 373, 1814–1823.
  14. Bedke, J.; Albiges, L.; Capitanio, U.; Giles, R.H.; Hora, M.; Lam, T.B.; Ljungberg, B.; Marconi, L.; Klatte, T.; Volpe, A.; et al. The 2021 Updated European Association of Urology Guidelines on Renal Cell Carcinoma: Immune Checkpoint Inhibitor-based Combination Therapies for Treatment-naive Metastatic Clear-cell Renal Cell Carcinoma Are Standard of Care. Eur. Urol. 2021, 80, 393–397.
  15. Zou, Z.; Tao, T.; Li, H.; Zhu, X. mTOR signaling pathway and mTOR inhibitors in cancer: Progress and challenges. Cell Biosci. 2020, 10, 31.
  16. Escudier, B.; Porta, C.; Schmidinger, M.; Rioux-Leclercq, N.; Bex, A.; Khoo, V.; Grünwald, V.; Gillessen, S.; Horwich, A. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann. Oncol. 2019, 30, 706–720.
  17. Tannir, N.M.; Msaouel, P.; Ross, J.A.; Devine, C.E.; Chandramohan, A.; Gonzalez, G.M.N.; Wang, X.; Wang, J.; Corn, P.G.; Lim, Z.D.; et al. Temsirolimus versus Pazopanib (TemPa) in Patients with Advanced Clear-cell Renal Cell Carcinoma and Poor-risk Features: A Randomized Phase II Trial. Eur. Urol. Oncol. 2020, 3, 687–694.
  18. Hudes, G.; Carducci, M.; Tomczak, P.; Dutcher, J.; Figlin, R.; Kapoor, A.; Staroslawska, E.; Sosman, J.; McDermott, D.; Bodrogi, I.; et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 2007, 356, 2271–2281.
  19. Motzer, R.J.; Escudier, B.; Oudard, S.; Hutson, T.E.; Porta, C.; Bracarda, S.; Grünwald, V.; Thompson, J.A.; Figlin, R.A.; Hollaender, N.; et al. Phase 3 trial of everolimus for metastatic renal cell carcinoma: Final results and analysis of prognostic factors. Cancer 2010, 116, 4256–4265.
  20. Motzer, R.J.; Hutson, T.E.; Ren, M.; Dutcus, C.; Larkin, J. Independent assessment of lenvatinib plus everolimus in patients with metastatic renal cell carcinoma. Lancet. Oncol. 2016, 17, e4–e5.
  21. Motzer, R.J.; Hutson, T.E.; Glen, H.; Michaelson, M.D.; Molina, A.; Eisen, T.; Jassem, J.; Zolnierek, J.; Maroto, J.P.; Mellado, B.; et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: A randomised, phase 2, open-label, multicentre trial. Lancet. Oncol. 2015, 16, 1473–1482.
  22. Motzer, R.J.; Escudier, B.; McDermott, D.F.; George, S.; Hammers, H.J.; Srinivas, S.; Tykodi, S.S.; Sosman, J.A.; Procopio, G.; Plimack, E.R.; et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2015, 373, 1803–1813.
  23. Cella, D.; Grünwald, V.; Nathan, P.; Doan, J.; Dastani, H.; Taylor, F.; Bennett, B.; DeRosa, M.; Berry, S.; Broglio, K.; et al. Quality of life in patients with advanced renal cell carcinoma given nivolumab versus everolimus in CheckMate 025: A randomised, open-label, phase 3 trial. Lancet. Oncol. 2016, 17, 994–1003.
  24. Quhal, F.; Mori, K.; Bruchbacher, A.; Resch, I.; Mostafaei, H.; Pradere, B.; Schuettfort, V.M.; Laukhtina, E.; Egawa, S.; Fajkovic, H.; et al. First-line Immunotherapy-based Combinations for Metastatic Renal Cell Carcinoma: A Systematic Review and Network Meta-analysis. Eur. Urol. Oncol. 2021, 4, 755–765.
  25. Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1103–1115.
  26. Albiges, L.; Tannir, N.M.; Burotto, M.; McDermott, D.; Plimack, E.R.; Barthelemy, P.; Porta, C.; Powles, T.; Donskov, F.; George, S.; et al. Nivolumab plus ipilimumab versus sunitinib for first-line treatment of advanced renal cell carcinoma: Extended 4-year follow-up of the phase III CheckMate 214 trial. ESMO Open 2020, 5, e001079.
  27. Yip, G.W.; Smollich, M.; Gotte, M. Therapeutic value of glycosaminoglycans in cancer. Mol. Cancer Ther. 2006, 5, 2139–2148.
  28. Afratis, N.; Gialeli, C.; Nikitovic, D.; Tsegenidis, T.; Karousou, E.; Theocharis, A.D.; Pavao, M.S.; Tzanakakis, G.N.; Karamanos, N.K. Glycosaminoglycans: Key players in cancer cell biology and treatment. FEBS J. 2012, 279, 1177–1197.
  29. Couto, M.R.; Rodrigues, J.L.; Rodrigues, L.R. Heterologous production of chondroitin. Biotechnol. Rep. 2022, 33, e00710.
  30. Essentials of Glycobiology , 4th ed.; Varki, A.; Cummings, R.D.; Esko, J.D.; Stanley, P.; Hart, G.W.; Aebi, M.; Mohnen, D.; Kinoshita, T.; Packer, N.H.; Prestegard, J.H.; et al. (Eds.) Cold Spring Harbor Laboratory Press: New York, NY, USA, 2022; Available online: https://www.ncbi.nlm.nih.gov/books/NBK579918/ (accessed on 9 November 2022).
  31. Vigetti, D.; Karousou, E.; Viola, M.; Deleonibus, S.; De Luca, G.; Passi, A. Hyaluronan: Biosynthesis and signaling. Biochim. Et Biophys. Acta 2014, 1840, 2452–2459.
  32. Gottschalk, J.; Elling, L. Current state on the enzymatic synthesis of glycosaminoglycans. Curr. Opin. Chem. Biol. 2021, 61, 71–80.
  33. Sudha, P.N.; Rose, M.H. Chapter Nine—Beneficial Effects of Hyaluronic Acid. In Advances in Food and Nutrition Research; Kim, S.-K., Ed.; Academic Press: Cambridge, MA, USA, 2014; Volume 72, pp. 137–176.
  34. Kogan, G.; Soltes, L.; Stern, R.; Gemeiner, P. Hyaluronic acid: A natural biopolymer with a broad range of biomedical and industrial applications. Biotechnol. Lett. 2007, 29, 17–25.
  35. Funderburgh, J.L. MINI REVIEW Keratan sulfate: Structure, biosynthesis, and function. Glycobiology 2000, 10, 951–958.
  36. Sasisekharan, R.; Shriver, Z.; Venkataraman, G.; Narayanasami, U. Roles of heparan-sulphate glycosaminoglycans in cancer. Nat. Rev. Cancer 2002, 2, 521–528.
  37. Hull, E.E.; Montgomery, M.R.; Leyva, K.J. Epigenetic Regulation of the Biosynthesis & Enzymatic Modification of Heparan Sulfate Proteoglycans: Implications for Tumorigenesis and Cancer Biomarkers. Int. J. Mol. Sci. 2017, 18, 1361.
  38. Koo, C.Y.; Bay, B.H.; Lui, P.C.; Tse, G.M.; Tan, P.H.; Yip, G.W. Immunohistochemical expression of heparan sulfate correlates with stromal cell proliferation in breast phyllodes tumors. Mod. Pathol. 2006, 19, 1344–1350.
  39. Mark, M.P.; Baker, J.R.; Kimata, K.; Ruch, J.V. Regulated changes in chondroitin sulfation during embryogenesis: An immunohistochemical approach. Int. J. Dev. Biol. 1990, 34, 191–204.
  40. Gama, C.I.; Tully, S.E.; Sotogaku, N.; Clark, P.M.; Rawat, M.; Vaidehi, N.; Goddard, W.A., 3rd; Nishi, A.; Hsieh-Wilson, L.C. Sulfation patterns of glycosaminoglycans encode molecular recognition and activity. Nat. Chem. Biol. 2006, 2, 467–473.
  41. Zou, X.H.; Foong, W.C.; Cao, T.; Bay, B.H.; Ouyang, H.W.; Yip, G.W. Chondroitin sulfate in palatal wound healing. J. Dent. Res. 2004, 83, 880–885.
  42. Lin, T.S.; Hsieh, C.H.; Kuo, C.; Juang, Y.P.; Hsieh, Y.S.Y.; Chiang, H.; Hung, S.C.; Jiang, C.C.; Liang, P.H. Sulfation pattern of chondroitin sulfate in human osteoarthritis cartilages reveals a lower level of chondroitin-4-sulfate. Carbohydr. Polym. 2020, 229, 115496.
  43. Kawamura, D.; Funakoshi, T.; Mizumoto, S.; Sugahara, K.; Iwasaki, N. Sulfation patterns of exogenous chondroitin sulfate affect chondrogenic differentiation of ATDC5 cells. J. Orthop. Sci. 2014, 19, 1028–1035.
  44. Lepucki, A.; Orlińska, K.; Mielczarek-Palacz, A.; Kabut, J.; Olczyk, P.; Komosińska-Vassev, K. The Role of Extracellular Matrix Proteins in Breast Cancer. J. Clin. Med. 2022, 11, 1250.
  45. Auvinen, P.; Tammi, R.; Parkkinen, J.; Tammi, M.; Agren, U.; Johansson, R.; Hirvikoski, P.; Eskelinen, M.; Kosma, V.M. Hyaluronan in peritumoral stroma and malignant cells associates with breast cancer spreading and predicts survival. Am. J. Pathol. 2000, 156, 529–536.
  46. Pirinen, R.; Tammi, R.; Tammi, M.; Hirvikoski, P.; Parkkinen, J.J.; Johansson, R.; Bohm, J.; Hollmen, S.; Kosma, V.M. Prognostic value of hyaluronan expression in non-small-cell lung cancer: Increased stromal expression indicates unfavorable outcome in patients with adenocarcinoma. Int. J. Cancer 2001, 95, 12–17.
  47. Anttila, M.A.; Tammi, R.H.; Tammi, M.I.; Syrjanen, K.J.; Saarikoski, S.V.; Kosma, V.M. High levels of stromal hyaluronan predict poor disease outcome in epithelial ovarian cancer. Cancer Res. 2000, 60, 150–155.
  48. Poh, Z.W.; Gan, C.H.; Lee, E.J.; Guo, S.; Yip, G.W.; Lam, Y. Divergent synthesis of chondroitin sulfate disaccharides and identification of sulfate motifs that inhibit triple negative breast cancer. Sci. Rep. 2015, 5, 14355.
  49. Kojima, J.; Nakamura, N.; Kanatani, M.; Omori, K. The glycosaminoglycans in human hepatic cancer. Cancer Res. 1975, 35, 542–547.
  50. Kovalszky, I.; Pogany, G.; Molnar, G.; Jeney, A.; Lapis, K.; Karacsonyi, S.; Szecseny, A.; Iozzo, R.V. Altered glycosaminoglycan composition in reactive and neoplastic human liver. Biochem. Biophys. Res. Commun. 1990, 167, 883–890.
  51. Horai, T.; Nakamura, N.; Tateishi, R.; Hattori, S. Glycosaminoglycans in human lung cancer. Cancer 1981, 48, 2016–2021.
  52. Theocharis, A.D.; Tsara, M.E.; Papageorgacopoulou, N.; Karavias, D.D.; Theocharis, D.A. Pancreatic carcinoma is characterized by elevated content of hyaluronan and chondroitin sulfate with altered disaccharide composition. Biochim. Et Biophys. Acta 2000, 1502, 201–206.
  53. Marolla, A.P.; Waisberg, J.; Saba, G.T.; Waisberg, D.R.; Margeotto, F.B.; Pinhal, M.A. Glycomics expression analysis of sulfated glycosaminoglycans of human colorectal cancer tissues and non-neoplastic mucosa by electrospray ionization mass spectrometry. Einstein (Sao Paulo) 2015, 13, 510–517.
  54. Sobue, M.; Takeuchi, J.; Miura, K.; Kawase, K.; Mizuno, F.; Sato, E. Glycosaminoglycan content and synthesis in gastric carcinoma. Br. J. Cancer 1980, 42, 78–84.
  55. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  56. Park, J.S.; Burckhardt, C.J.; Lazcano, R.; Solis, L.M.; Isogai, T.; Li, L.; Chen, C.S.; Gao, B.; Minna, J.D.; Bachoo, R.; et al. Mechanical regulation of glycolysis via cytoskeleton architecture. Nature 2020, 578, 621–626.
  57. Ahmadzadeh, H.; Webster, M.R.; Behera, R.; Jimenez Valencia, A.M.; Wirtz, D.; Weeraratna, A.T.; Shenoy, V.B. Modeling the two-way feedback between contractility and matrix realignment reveals a nonlinear mode of cancer cell invasion. Proc. Natl. Acad. Sci. USA 2017, 114, E1617–E1626.
  58. Kaur, A.; Ecker, B.L.; Douglass, S.M.; Kugel, C.H., 3rd; Webster, M.R.; Almeida, F.V.; Somasundaram, R.; Hayden, J.; Ban, E.; Ahmadzadeh, H.; et al. Remodeling of the Collagen Matrix in Aging Skin Promotes Melanoma Metastasis and Affects Immune Cell Motility. Cancer Discov. 2019, 9, 64–81.
  59. Mouw, J.K.; Ou, G.; Weaver, V.M. Extracellular matrix assembly: A multiscale deconstruction. Nat. Rev. Mol. Cell Biol. 2014, 15, 771–785.
  60. Baghy, K.; Tátrai, P.; Regős, E.; Kovalszky, I. Proteoglycans in liver cancer. World J. Gastroenterol. 2016, 22, 379–393.
  61. Kosaki, R.; Watanabe, K.; Yamaguchi, Y. Overproduction of hyaluronan by expression of the hyaluronan synthase Has2 enhances anchorage-independent growth and tumorigenicity. Cancer Res. 1999, 59, 1141–1145.
  62. Itano, N.; Sawai, T.; Miyaishi, O.; Kimata, K. Relationship between hyaluronan production and metastatic potential of mouse mammary carcinoma cells. Cancer Res. 1999, 59, 2499–2504.
  63. Liu, N.; Gao, F.; Han, Z.; Xu, X.; Underhill, C.B.; Zhang, L. Hyaluronan synthase 3 overexpression promotes the growth of TSU prostate cancer cells. Cancer Res. 2001, 61, 5207–5214.
  64. Jacobson, A.; Rahmanian, M.; Rubin, K.; Heldin, P. Expression of hyaluronan synthase 2 or hyaluronidase 1 differentially affect the growth rate of transplantable colon carcinoma cell tumors. Int. J. Cancer 2002, 102, 212–219.
  65. Toole, B.P. Hyaluronan in morphogenesis. Semin. Cell Dev. Biol. 2001, 12, 79–87.
  66. Evanko, S.P.; Angello, J.C.; Wight, T.N. Formation of hyaluronan- and versican-rich pericellular matrix is required for proliferation and migration of vascular smooth muscle cells. Arter. Thromb. Vasc. Biol. 1999, 19, 1004–1013.
  67. Hayen, W.; Goebeler, M.; Kumar, S.; Riessen, R.; Nehls, V. Hyaluronan stimulates tumor cell migration by modulating the fibrin fiber architecture. J. Cell Sci. 1999, 112 Pt 13, 2241–2251.
  68. Simpson, M.A.; Wilson, C.M.; Furcht, L.T.; Spicer, A.P.; Oegema, T.R., Jr.; McCarthy, J.B. Manipulation of hyaluronan synthase expression in prostate adenocarcinoma cells alters pericellular matrix retention and adhesion to bone marrow endothelial cells. J. Biol. Chem. 2002, 277, 10050–10057.
  69. Offeddu, G.S.; Hajal, C.; Foley, C.R.; Wan, Z.; Ibrahim, L.; Coughlin, M.F.; Kamm, R.D. The cancer glycocalyx mediates intravascular adhesion and extravasation during metastatic dissemination. Commun. Biol. 2021, 4, 255.
  70. Turley, E.A.; Noble, P.W.; Bourguignon, L.Y. Signaling properties of hyaluronan receptors. J. Biol. Chem. 2002, 277, 4589–4592.
  71. Day, A.J.; Prestwich, G.D. Hyaluronan-binding proteins: Tying up the giant. J. Biol. Chem. 2002, 277, 4585–4588.
  72. Yu, Q.; Toole, B.P.; Stamenkovic, I. Induction of apoptosis of metastatic mammary carcinoma cells in vivo by disruption of tumor cell surface CD44 function. J. Exp. Med. 1997, 186, 1985–1996.
  73. Misra, S.; Hascall, V.C.; Markwald, R.R.; Ghatak, S. Interactions between Hyaluronan and Its Receptors (CD44, RHAMM) Regulate the Activities of Inflammation and Cancer. Front. Immunol. 2015, 6, 201.
  74. Mohapatra, S.; Yang, X.; Wright, J.A.; Turley, E.A.; Greenberg, A.H. Soluble hyaluronan receptor RHAMM induces mitotic arrest by suppressing Cdc2 and cyclin B1 expression. J. Exp. Med. 1996, 183, 1663–1668.
  75. Zhang, Y.; Thant, A.A.; Machida, K.; Ichigotani, Y.; Naito, Y.; Hiraiwa, Y.; Senga, T.; Sohara, Y.; Matsuda, S.; Hamaguchi, M. Hyaluronan-CD44s signaling regulates matrix metalloproteinase-2 secretion in a human lung carcinoma cell line QG90. Cancer Res. 2002, 62, 3962–3965.
  76. Chambers, A.F.; Matrisian, L.M. Changing views of the role of matrix metalloproteinases in metastasis. J. Natl. Cancer Inst. 1997, 89, 1260–1270.
  77. Li, M.; Yan, T.; Cai, Y.; Wei, Y.; Xie, Q. Expression of matrix metalloproteinases and their association with clinical characteristics of solid tumors. Gene 2022, 850, 146927.
  78. Hall, C.L.; Wang, C.; Lange, L.A.; Turley, E.A. Hyaluronan and the hyaluronan receptor RHAMM promote focal adhesion turnover and transient tyrosine kinase activity. J. Cell Biol. 1994, 126, 575–588.
  79. Assmann, V.; Jenkinson, D.; Marshall, J.F.; Hart, I.R. The intracellular hyaluronan receptor RHAMM/IHABP interacts with microtubules and actin filaments. J. Cell Sci. 1999, 112 Pt 22, 3943–3954.
  80. Maxwell, C.A.; Keats, J.J.; Crainie, M.; Sun, X.; Yen, T.; Shibuya, E.; Hendzel, M.; Chan, G.; Pilarski, L.M. RHAMM is a centrosomal protein that interacts with dynein and maintains spindle pole stability. Mol. Biol. Cell 2003, 14, 2262–2276.
  81. Wu, H.J.; Hao, M.; Yeo, S.K.; Guan, J.L. FAK signaling in cancer-associated fibroblasts promotes breast cancer cell migration and metastasis by exosomal miRNAs-mediated intercellular communication. Oncogene 2020, 39, 2539–2549.
  82. Iozzo, R.V.; Schaefer, L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol. J. Int. Soc. Matrix Biol. 2015, 42, 11–55.
  83. Pudełko, A.; Wisowski, G.; Olczyk, K.; Koźma, E.M. The dual role of the glycosaminoglycan chondroitin-6-sulfate in the development, progression and metastasis of cancer. FEBS J. 2019, 286, 1815–1837.
  84. Kieber-Emmons, A.M.; Jousheghany, F.; Monzavi-Karbassi, B. On the role of cell surface chondroitin sulfates and their core proteins in breast cancer metastasis. In Breast Cancer—Focusing Tumor Microenvironment, Stem Cells and Metastasis; Gunduz, M., Gunduz, E., Eds.; InTech: London, UK, 2011; pp. 435–450.
  85. Alini, M.; Losa, G.A. Partial characterization of proteoglycans isolated from neoplastic and nonneoplastic human breast tissues. Cancer Res. 1991, 51, 1443–1447.
  86. Olsen, E.B.; Trier, K.; Eldov, K.; Ammitzboll, T. Glycosaminoglycans in human breast cancer. Acta Obstet. Et Gynecol. Scand. 1988, 67, 539–542.
  87. Nadanaka, S.; Kinouchi, H.; Kitagawa, H. Chondroitin sulfate-mediated N-cadherin/β-catenin signaling is associated with basal-like breast cancer cell invasion. J. Biol. Chem. 2018, 293, 444–465.
  88. Ricciardelli, C.; Brooks, J.H.; Suwiwat, S.; Sakko, A.J.; Mayne, K.; Raymond, W.A.; Seshadri, R.; LeBaron, R.G.; Horsfall, D.J. Regulation of stromal versican expression by breast cancer cells and importance to relapse-free survival in patients with node-negative primary breast cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2002, 8, 1054–1060.
  89. Suwiwat, S.; Ricciardelli, C.; Tammi, R.; Tammi, M.; Auvinen, P.; Kosma, V.M.; LeBaron, R.G.; Raymond, W.A.; Tilley, W.D.; Horsfall, D.J. Expression of extracellular matrix components versican, chondroitin sulfate, tenascin, and hyaluronan, and their association with disease outcome in node-negative breast cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2004, 10, 2491–2498.
  90. Teng, Y.H.; Tan, P.H.; Chia, S.J.; Zam, N.A.; Lau, W.K.; Cheng, C.W.; Bay, B.H.; Yip, G.W. Increased expression of non-sulfated chondroitin correlates with adverse clinicopathological parameters in prostate cancer. Mod. Pathol. 2008, 21, 893–901.
  91. Willis, C.M.; Klüppel, M. Chondroitin sulfate-E is a negative regulator of a pro-tumorigenic Wnt/beta-catenin-Collagen 1 axis in breast cancer cells. PLoS ONE 2014, 9, e103966.
  92. Troeberg, L.; Lazenbatt, C.; Anower, E.K.M.F.; Freeman, C.; Federov, O.; Habuchi, H.; Habuchi, O.; Kimata, K.; Nagase, H. Sulfated glycosaminoglycans control the extracellular trafficking and the activity of the metalloprotease inhibitor TIMP-3. Chem. Biol. 2014, 21, 1300–1309.
  93. ten Dam, G.B.; van de Westerlo, E.M.; Purushothaman, A.; Stan, R.V.; Bulten, J.; Sweep, F.C.; Massuger, L.F.; Sugahara, K.; van Kuppevelt, T.H. Antibody GD3G7 selected against embryonic glycosaminoglycans defines chondroitin sulfate-E domains highly up-regulated in ovarian cancer and involved in vascular endothelial growth factor binding. Am. J. Pathol. 2007, 171, 1324–1333.
  94. Mizumoto, S.; Fongmoon, D.; Sugahara, K. Interaction of chondroitin sulfate and dermatan sulfate from various biological sources with heparin-binding growth factors and cytokines. Glycoconj. J. 2013, 30, 619–632.
  95. Monzavi-Karbassi, B.; Stanley, J.S.; Hennings, L.; Jousheghany, F.; Artaud, C.; Shaaf, S.; Kieber-Emmons, T. Chondroitin sulfate glycosaminoglycans as major P-selectin ligands on metastatic breast cancer cell lines. Int. J. Cancer 2007, 120, 1179–1191.
  96. Murai, T.; Sougawa, N.; Kawashima, H.; Yamaguchi, K.; Miyasaka, M. CD44-chondroitin sulfate interactions mediate leukocyte rolling under physiological flow conditions. Immunol. Lett. 2004, 93, 163–170.
  97. Sage, J.; Mallèvre, F.; Barbarin-Costes, F.; Samsonov, S.A.; Gehrcke, J.P.; Pisabarro, M.T.; Perrier, E.; Schnebert, S.; Roget, A.; Livache, T.; et al. Binding of chondroitin 4-sulfate to cathepsin S regulates its enzymatic activity. Biochemistry 2013, 52, 6487–6498.
  98. Olson, O.C.; Joyce, J.A. Cysteine cathepsin proteases: Regulators of cancer progression and therapeutic response. Nat. Rev. Cancer 2015, 15, 712–729.
  99. Zhangyuan, G.; Wang, F.; Zhang, H.; Jiang, R.; Tao, X.; Yu, D.; Jin, K.; Yu, W.; Liu, Y.; Yin, Y.; et al. VersicanV1 promotes proliferation and metastasis of hepatocellular carcinoma through the activation of EGFR-PI3K-AKT pathway. Oncogene 2020, 39, 1213–1230.
  100. Zhang, Y.; Zou, X.; Qian, W.; Weng, X.; Zhang, L.; Zhang, L.; Wang, S.; Cao, X.; Ma, L.; Wei, G.; et al. Enhanced PAPSS2/VCAN sulfation axis is essential for Snail-mediated breast cancer cell migration and metastasis. Cell Death Differ. 2019, 26, 565–579.
  101. Yeung, T.L.; Leung, C.S.; Wong, K.K.; Samimi, G.; Thompson, M.S.; Liu, J.; Zaid, T.M.; Ghosh, S.; Birrer, M.J.; Mok, S.C. TGF-β modulates ovarian cancer invasion by upregulating CAF-derived versican in the tumor microenvironment. Cancer Res. 2013, 73, 5016–5028.
  102. Guo, J.Y.; Hsu, H.S.; Tyan, S.W.; Li, F.Y.; Shew, J.Y.; Lee, W.H.; Chen, J.Y. Serglycin in tumor microenvironment promotes non-small cell lung cancer aggressiveness in a CD44-dependent manner. Oncogene 2017, 36, 2457–2471.
  103. Guo, J.Y.; Chiu, C.H.; Wang, M.J.; Li, F.A.; Chen, J.Y. Proteoglycan serglycin promotes non-small cell lung cancer cell migration through the interaction of its glycosaminoglycans with CD44. J. Biomed. Sci. 2020, 27, 2.
  104. Bouris, P.; Manou, D.; Sopaki-Valalaki, A.; Kolokotroni, A.; Moustakas, A.; Kapoor, A.; Iozzo, R.V.; Karamanos, N.K.; Theocharis, A.D. Serglycin promotes breast cancer cell aggressiveness: Induction of epithelial to mesenchymal transition, proteolytic activity and IL-8 signaling. Matrix Biol. J. Int. Soc. Matrix Biol. 2018, 74, 35–51.
  105. Itano, N.; Atsumi, F.; Sawai, T.; Yamada, Y.; Miyaishi, O.; Senga, T.; Hamaguchi, M.; Kimata, K. Abnormal accumulation of hyaluronan matrix diminishes contact inhibition of cell growth and promotes cell migration. Proc. Natl. Acad. Sci. USA 2002, 99, 3609–3614.
  106. Iravani, O.; Bay, B.H.; Yip, G.W. Silencing HS6ST3 inhibits growth and progression of breast cancer cells through suppressing IGF1R and inducing XAF1. Exp. Cell Res. 2017, 350, 380–389.
  107. Vijaya Kumar, A.; Salem Gassar, E.; Spillmann, D.; Stock, C.; Sen, Y.P.; Zhang, T.; Van Kuppevelt, T.H.; Hulsewig, C.; Koszlowski, E.O.; Pavao, M.S.; et al. HS3ST2 modulates breast cancer cell invasiveness via MAP kinase- and Tcf4 (Tcf7l2)-dependent regulation of protease and cadherin expression. Int. J. Cancer 2014, 135, 2579–2592.
  108. Kumagai, S.; Ishibashi, K.; Kataoka, M.; Oguro, T.; Kiko, Y.; Yanagida, T.; Aikawa, K.; Kojima, Y. Impact of Sulfatase-2 on cancer progression and prognosis in patients with renal cell carcinoma. Cancer Sci. 2016, 107, 1632–1641.
  109. Song, K.; Li, Q.; Jiang, Z.Z.; Guo, C.W.; Li, P. Heparan sulfate D-glucosaminyl 3-O-sulfotransferase-3B1, a novel epithelial-mesenchymal transition inducer in pancreatic cancer. Cancer Biol. Ther. 2011, 12, 388–398.
  110. Ibrahim, S.A.; Gadalla, R.; El-Ghonaimy, E.A.; Samir, O.; Mohamed, H.T.; Hassan, H.; Greve, B.; El-Shinawi, M.; Mohamed, M.M.; Gotte, M. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways. Mol. Cancer 2017, 16, 57.
  111. Kumar Katakam, S.; Tria, V.; Sim, W.C.; Yip, G.W.; Molgora, S.; Karnavas, T.; Elghonaimy, E.A.; Pelucchi, P.; Piscitelli, E.; Ibrahim, S.A.; et al. The heparan sulfate proteoglycan syndecan-1 regulates colon cancer stem cell function via a focal adhesion kinase-Wnt signaling axis. FEBS J. 2021, 288, 486–506.
  112. Grindel, B.J.; Martinez, J.R.; Tellman, T.V.; Harrington, D.A.; Zafar, H.; Nakhleh, L.; Chung, L.W.; Farach-Carson, M.C. Matrilysin/MMP-7 Cleavage of Perlecan/HSPG2 Complexed with Semaphorin 3A Supports FAK-Mediated Stromal Invasion by Prostate Cancer Cells. Sci. Rep. 2018, 8, 7262.
  113. Leiphrakpam, P.D.; Patil, P.P.; Remmers, N.; Swanson, B.; Grandgenett, P.M.; Qiu, F.; Yu, F.; Radhakrishnan, P. Role of keratan sulfate expression in human pancreatic cancer malignancy. Sci. Rep. 2019, 9, 9665.
  114. Yang, C.T.; Li, J.M.; Chu, W.K.; Chow, S.E. Downregulation of lumican accelerates lung cancer cell invasion through p120 catenin. Cell Death Dis. 2018, 9, 414.
  115. Lu, Y.P.; Ishiwata, T.; Kawahara, K.; Watanabe, M.; Naito, Z.; Moriyama, Y.; Sugisaki, Y.; Asano, G. Expression of lumican in human colorectal cancer cells. Pathol. Int. 2002, 52, 519–526.
  116. Radwanska, A.; Litwin, M.; Nowak, D.; Baczynska, D.; Wegrowski, Y.; Maquart, F.X.; Malicka-Blaszkiewicz, M. Overexpression of lumican affects the migration of human colon cancer cells through up-regulation of gelsolin and filamentous actin reorganization. Exp. Cell Res. 2012, 318, 2312–2323.
More
Information
Subjects: Oncology; Cell Biology
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: 339
Revisions: 2 times (View History)
Update Date: 17 Jan 2023
1000/1000
Video Production Service