Next Article in Journal
Dichloroacetyl Amides of 3,5-Bis(benzylidene)-4-piperidones Displaying Greater Toxicity to Neoplasms than to Non-Malignant Cells
Previous Article in Journal
Osteopathic Manipulative Medicine: A Brief Review of the Hands-On Treatment Approaches and Their Therapeutic Uses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Glycobiology of Cancer: Sugar Drives the Show

by
Jhenifer Santos dos Reis
1,
Marcos André Rodrigues da Costa Santos
1,
Daniella Pereira Mendonça
1,
Stefani Ingrid Martins do Nascimento
1,
Pedro Marçal Barcelos
1,
Rafaela Gomes Correia de Lima
1,
Kelli Monteiro da Costa
1,
Celio Geraldo Freire-de-Lima
1,
Alexandre Morrot
2,3,
Jose Osvaldo Previato
1,
Lucia Mendonça Previato
1,
Leonardo Marques da Fonseca
1 and
Leonardo Freire-de-Lima
1,*
1
Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil
2
Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, Brazil
3
Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21044-020, Brazil
*
Author to whom correspondence should be addressed.
Submission received: 26 April 2022 / Revised: 19 May 2022 / Accepted: 20 May 2022 / Published: 24 May 2022

Abstract

:
Cancer development and progression is associated with aberrant changes in cellular glycosylation. Cells expressing altered glycan-structures are recognized by cells of the immune system, favoring the induction of inhibitory immune processes which subsequently promote tumor growth and spreading. Here, we discuss about the importance of glycobiology in modern medicine, taking into account the impact of altered glycan structures expressed in cancer cells as potential glycobiomarkers of disease, as well as on cancer development and progression.

The hottest topic in the field of molecular biology during the 1970s and 80s was understanding the flow of information solely between DNA, RNA and proteins [1]. The central dogma of molecular biology states that once that information takes the form of proteins, it cannot be taken back to nucleic acid [2]. Although disproved by the discovery of prion-mediated heredity, its reflections can still be perceived to this day [3]. Clear examples are the illustrations of cell membranes in classical textbooks of biochemistry and cell and molecular biology, where little is discussed about the importance of glycoconjugates. Thanks to scientific and technological advances, nowadays it is well accepted that cell-surface and/or secreted glycomes reflect overall cellular status in health and disease [4]. The term glycome refers to the complete repertoire of glycomolecules decorated with carbohydrate chains, or glycans, that are covalently linked to lipid or proteins [5]. Glycosylation is a highly dynamic and finely regulated process involving a complex biological apparatus whose components are spread into different cellular compartments, such as nucleus, cytoplasm, endoplasmic reticulum, Golgi and lysosomes [6,7,8]. It is estimated that 3–4% of the human genome encodes elements of the glycoconjugate biosynthesis machinery. Among such components we can find enzymes, which are generically called glycosyltransferases and glycosidases, chaperones, sugar transporters and donors, as well as other molecules necessary for the modification of proteins or lipids with carbohydrates [8].
Glycoconjugates are found on the cell surface of all living organisms, and play essential roles in mediating protein-receptor signaling, cell–cell and cell–matrix interactions, and appropriate protein folding and maturation during translation [9]. In fact, changes in glycosylation can modulate inflammatory responses, enable viral immune escape, promote cancer cell metastasis or regulate apoptosis [10]. New insights into the structure and function of the glycome can now be applied to therapy development and could improve our ability to fine-tune immunological responses and inflammation, optimize the performance of therapeutic antibodies and boost immune responses to chronic diseases, such as cancer [11,12]. These examples illustrate the potential of the emerging field of glycomedicine, which communally aim to clarify the function of glycans in person-to-person and between-population discrepancies in disease vulnerability and response to health interventions such as vaccines, nutrition and drugs [13].
Targeting immune checkpoints to improve the outcome of cancer patients is an ongoing discussion in oncobiology. However, few patients have shown long-term benefits from currently used CTLA-4 and PD-1/PD-L1 inhibitors [14,15,16]. Therefore, new strategies are needed to increase the long-term remission after cancer immunotherapy. Over the past ten years, numerous studies have shown that glyco-immune checkpoints can be used as new targets for cancer immunotherapy. They are well-defined as immunomodulatory pathways, including interactions between glycan-binding proteins or lectins with glycan epitopes [14,17,18]. The most prominent pathways involve the immune and vascular programs triggered by galectins [19,20,21,22], as well as the sialo-glycan-Siglec axis [14,23,24,25]. In both cases, inhibitors are already being successfully tested in clinical trials [19,26]. This confirms that advances in the field of glyco-immunology will permit us to improve cancer immunotherapy and help many patients.
Regarding the effects of glycoconjugates in cancer cells, it has been well documented that malignant transformation and tumor progression correlate with aberrant changes in cellular glycosylation [6,12,27]. In cancer cells, O-linked glycans are characterized to present immature and/or truncated structures due to reduced expression and/or activity of specific glycosyltransferases, such as beta 1,3-glalactosyltransferase [28] and core 2 beta-1,6-N-acetylglucosaminyltransferase (C2GNT), contributing to the accumulation of altered glycan structures such as Tn and sialyl-Tn antigens [29] and T-antigen and T-sialyl antigen [30]. In contrast, N-linked glycans in cancer cells are characterized by being long, branched, and hypersialylated [30]. When it comes to N-linked glycans, however, there is more to the story. Many groups show an abundance of long, branched, and hypersialylated structures [30,31,32,33,34,35], while others report high mannose structures [36,37] or even both [38]. One particular study points to high mannose structures being prevalent in the primary tumor, while branched sialylated epitopes are found in metastatic foci [39]. These findings may suggest that just like it’s very hard to find two cancer patients suffering from the exact same disease, glycosylation patterns may vary depending on the precise mutations occurring simultaneously on the cancer cell.
For a long time, such structures were used only for diagnostic purposes. However, many research groups have since demonstrated that structurally altered glycoproteins are able to modulate various events linked to the progression of different types of cancer [40]. Recent studies have demonstrated that altered glycosylation of proteins that make up the glycocalyx may be recognized by immune cells, leading to induction of inhibitory immune processes, which subsequently drive tumor growth and metastasis [41]. Several studies developed by our research group demonstrated that O- and N-linked unusual glycan structures govern phenomena associated to the epithelial–mesenchymal transition (EMT) process, as well as the acquisition/maintenance of the multidrug resistance (MDR) phenotype [12,34,35,42,43,44,45,46,47,48,49,50]. MDR phenotype and the acquisition of metastatic properties by cancer cells are known as the main obstacles to the treatment of different types of cancer [51]. Over twenty-five years ago, these events were studied as independent phenomena. However, it is now well established that both are necessary to be investigated together [35], since numerous papers have demonstrated that glycan structures present strong impact on the MDR phenotype [12,52,53]. Although several studies have already described that the emergence of aberrant glycan structures is strictly related with the activation of both molecular pathways linked to EMT process and the emergence of MDR phenotype, little is known about how such glycan structures might connect these two multifactorial events. The developing of glycosyltransferase knockouts mice has confirmed that pathological phenotypes may be triggered in vivo by genetic manipulation of glycans [54], demonstrating that proteins decorated with aberrant glycan structures are promising drug targets for treating various diseases, including cancer.
In recent works, we have demonstrated for the first time that alterations in glycosylation in tumor cells chronically exposed to chemotherapeutic agents, are able to connect both MDR phenotype and EMT process, since in addition to presenting changes in the expression and/or activity of efflux pumps belonging to the ABC superfamily (ABCB1, ABCC1 and ABCG2) [55,56], the chemoresistant human cancer cell lines also showed increased cell motility, as well as altered expression of epithelial–mesenchymal markers, when compared with their normal counterparts [34,35]. These findings confirm the idea that both accretion of MDR phenotype and the activation of EMT process, which have been considered indispensable for invasion and metastasis [57,58,59], are deeply linked with unusual glycan structures expressed by transformed cells. In our previous study we also observed that the chronic exposure to non-lethal concentrations of cisplatin induced the expression of an isoform of fibronectin (FN), so called oncofetal FN (onf-FN) [35], which may be found in transformed cells, and embryonic samples, but is absent in normal tissues [6]. onf-FN was also described by Hakomori’s group in cancer cells undergoing EMT [42,43], but its role in many events linked to cancer progression, including the acquisition of drug-resistant phenotype, is still unknown. Taken together, it has become clear that further investigation in this area may offer new diagnostic biomarkers and therapeutic targets to combat this devastating disease, which while no longer a death sentence, is still considered potentially fatal if not diagnosed early.

Author Contributions

Conceptualization, J.S.d.R., L.F.-d.-L., L.M.d.F., D.P.M., S.I.M.d.N., P.M.B. and M.A.R.d.C.S.; manuscript writing, J.S.d.R., L.F.-d.-L., L.M.d.F., D.P.M., R.G.C.d.L., S.I.M.d.N., P.M.B., K.M.d.C. and M.A.R.d.C.S.; supervision, C.G.F.-d.-L., A.M., L.M.P., J.O.P. and L.F.-d.-L.; project administration, L.F.-d.-L. and J.S.d.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Brazilian National Research Council (CNPq), Brazilian Cancer Foundation, Rio de Janeiro State Science Foundation (FAPERJ).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Piras, V.; Tomita, M.; Selvarajoo, K. Is central dogma a global property of cellular information flow? Front. Physiol. 2012, 3, 439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Crick, F. Central dogma of molecular biology. Nature 1970, 227, 561–563. [Google Scholar] [CrossRef] [PubMed]
  3. Koonin, E.V. Does the central dogma still stand? Biol. Direct 2012, 7, 27. [Google Scholar] [CrossRef] [Green Version]
  4. Broussard, A.C.; Boyce, M. Life is sweet: The cell biology of glycoconjugates. Mol. Biol. Cell 2019, 30, 525–529. [Google Scholar] [CrossRef] [PubMed]
  5. Reily, C.; Stewart, T.J.; Renfrow, M.B.; Novak, J. Glycosylation in health and disease. Nat. Rev. Nephrol. 2019, 15, 346–366. [Google Scholar] [CrossRef] [PubMed]
  6. Freire-de-Lima, L. Sweet and sour: The impact of differential glycosylation in cancer cells undergoing epithelial-mesenchymal transition. Front. Oncol. 2014, 4, 59. [Google Scholar] [CrossRef] [Green Version]
  7. Bindeman, W.E.; Fingleton, B. Glycosylation as a regulator of site-specific metastasis. Cancer Metastasis Rev. 2022, 41, 107–129. [Google Scholar] [CrossRef]
  8. Schjoldager, K.T.; Narimatsu, Y.; Joshi, H.J.; Clausen, H. Global view of human protein glycosylation pathways and functions. Nat. Rev. Mol. Cell Biol. 2020, 21, 729–749. [Google Scholar] [CrossRef]
  9. Spiro, R.G. Protein glycosylation: Nature, distribution, enzymatic formation, and disease implications of glycopeptide bonds. Glycobiology 2002, 12, 43R–56R. [Google Scholar] [CrossRef]
  10. Groux-Degroote, S.; Cavdarli, S.; Uchimura, K.; Allain, F.; Delannoy, P. Glycosylation changes in inflammatory diseases. Adv. Protein Chem. Struct. Biol. 2020, 119, 111–156. [Google Scholar] [CrossRef]
  11. Scott, D.A.; Drake, R.R. Glycosylation and its implications in breast cancer. Expert Rev. Proteom. 2019, 16, 665–680. [Google Scholar] [CrossRef] [PubMed]
  12. Da Fonseca, L.M.; da Silva, V.A.; Freire-de-Lima, L.; Previato, J.O.; Mendonca-Previato, L.; Capella, M.A. Glycosylation in Cancer: Interplay between Multidrug Resistance and Epithelial-to-Mesenchymal Transition? Front. Oncol. 2016, 6, 158. [Google Scholar] [CrossRef] [PubMed]
  13. Kunej, T. Rise of Systems Glycobiology and Personalized Glycomedicine: Why and How to Integrate Glycomics with Multiomics Science? OMICS 2019, 23, 615–622. [Google Scholar] [CrossRef] [Green Version]
  14. Manni, M.; Laubli, H. Targeting glyco-immune checkpoints for cancer therapy. Expert Opin. Biol. Ther. 2021, 21, 1063–1071. [Google Scholar] [CrossRef]
  15. Wojtukiewicz, M.Z.; Rek, M.M.; Karpowicz, K.; Gorska, M.; Politynska, B.; Wojtukiewicz, A.M.; Moniuszko, M.; Radziwon, P.; Tucker, S.C.; Honn, K.V. Inhibitors of immune checkpoints-PD-1, PD-L1, CTLA-4-new opportunities for cancer patients and a new challenge for internists and general practitioners. Cancer Metastasis Rev. 2021, 40, 949–982. [Google Scholar] [CrossRef] [PubMed]
  16. Pandey, P.; Khan, F.; Qari, H.A.; Upadhyay, T.K.; Alkhateeb, A.F.; Oves, M. Revolutionization in Cancer Therapeutics via Targeting Major Immune Checkpoints PD-1, PD-L1 and CTLA-4. Pharmaceuticals 2022, 15, 335. [Google Scholar] [CrossRef]
  17. Wang, J.; Manni, M.; Barenwaldt, A.; Wieboldt, R.; Kirchhammer, N.; Ivanek, R.; Stanczak, M.; Zippelius, A.; Konig, D.; Rodrigues Manutano, N.; et al. Siglec Receptors Modulate Dendritic Cell Activation and Antigen Presentation to T Cells in Cancer. Front. Cell Dev. Biol. 2022, 10, 828916. [Google Scholar] [CrossRef]
  18. Chiang, A.W.T.; Baghdassarian, H.M.; Kellman, B.P.; Bao, B.; Sorrentino, J.T.; Liang, C.; Kuo, C.C.; Masson, H.O.; Lewis, N.E. Systems glycobiology for discovering drug targets, biomarkers, and rational designs for glyco-immunotherapy. J. Biomed. Sci. 2021, 28, 50. [Google Scholar] [CrossRef]
  19. Compagno, D.; Tiraboschi, C.; Garcia, J.D.; Rondon, Y.; Corapi, E.; Velazquez, C.; Laderach, D.J. Galectins as Checkpoints of the Immune System in Cancers, Their Clinical Relevance, and Implication in Clinical Trials. Biomolecules 2020, 10, 750. [Google Scholar] [CrossRef]
  20. Videla-Richardson, G.A.; Morris-Hanon, O.; Torres, N.I.; Esquivel, M.I.; Vera, M.B.; Ripari, L.B.; Croci, D.O.; Sevlever, G.E.; Rabinovich, G.A. Galectins as Emerging Glyco-Checkpoints and Therapeutic Targets in Glioblastoma. Int. J. Mol. Sci. 2021, 23, 316. [Google Scholar] [CrossRef]
  21. Mendez-Huergo, S.P.; Blidner, A.G.; Rabinovich, G.A. Galectins: Emerging regulatory checkpoints linking tumor immunity and angiogenesis. Curr. Opin. Immunol. 2017, 45, 8–15. [Google Scholar] [CrossRef] [PubMed]
  22. Sundblad, V.; Morosi, L.G.; Geffner, J.R.; Rabinovich, G.A. Galectin-1: A Jack-of-All-Trades in the Resolution of Acute and Chronic Inflammation. J. Immunol. 2017, 199, 3721–3730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wielgat, P.; Rogowski, K.; Niemirowicz-Laskowska, K.; Car, H. Sialic Acid-Siglec Axis as Molecular Checkpoints Targeting of Immune System: Smart Players in Pathology and Conventional Therapy. Int. J. Mol. Sci. 2020, 21, 4361. [Google Scholar] [CrossRef] [PubMed]
  24. Barenwaldt, A.; Laubli, H. The sialoglycan-Siglec glyco-immune checkpoint—A target for improving innate and adaptive anti-cancer immunity. Expert Opin. Ther. Targets 2019, 23, 839–853. [Google Scholar] [CrossRef] [PubMed]
  25. Wielgat, P.; Czarnomysy, R.; Trofimiuk, E.; Car, H. The sialoglycan-Siglec-E checkpoint axis in dexamethasone-induced immune subversion in glioma-microglia transwell co-culture system. Immunol. Res. 2019, 67, 348–357. [Google Scholar] [CrossRef]
  26. Lenza, M.P.; Atxabal, U.; Oyenarte, I.; Jimenez-Barbero, J.; Ereno-Orbea, J. Current Status on Therapeutic Molecules Targeting Siglec Receptors. Cells 2020, 9, 2691. [Google Scholar] [CrossRef]
  27. Nardy, A.F.; Freire-de-Lima, L.; Freire-de-Lima, C.G.; Morrot, A. The Sweet Side of Immune Evasion: Role of Glycans in the Mechanisms of Cancer Progression. Front. Oncol. 2016, 6, 54. [Google Scholar] [CrossRef] [Green Version]
  28. Xia, T.; Xiang, T.; Xie, H. Update on the role of C1GALT1 in cancer. Oncol. Lett. 2022, 23, 97. [Google Scholar] [CrossRef]
  29. Ju, T.; Wang, Y.; Aryal, R.P.; Lehoux, S.D.; Ding, X.; Kudelka, M.R.; Cutler, C.; Zeng, J.; Wang, J.; Sun, X.; et al. Tn and sialyl-Tn antigens, aberrant O-glycomics as human disease markers. Proteom. Clin. Appl. 2013, 7, 618–631. [Google Scholar] [CrossRef] [Green Version]
  30. Mereiter, S.; Balmana, M.; Campos, D.; Gomes, J.; Reis, C.A. Glycosylation in the Era of Cancer-Targeted Therapy: Where Are We Heading? Cancer Cell 2019, 36, 6–16. [Google Scholar] [CrossRef]
  31. Wang, L.; Yang, L.; Zhang, Y.; Lu, H. Dual isotopic labeling combined with fluorous solid-phase extraction for simultaneous discovery of neutral/sialylated N-glycans as biomarkers for gastric cancer. Anal. Chim. Acta 2020, 1104, 87–94. [Google Scholar] [CrossRef]
  32. Peng, W.; Zhu, R.; Zhou, S.; Mirzaei, P.; Mechref, Y. Integrated Transcriptomics, Proteomics, and Glycomics Reveals the Association between Up-regulation of Sialylated N-glycans/Integrin and Breast Cancer Brain Metastasis. Sci. Rep. 2019, 9, 17361. [Google Scholar] [CrossRef]
  33. McDowell, C.T.; Klamer, Z.; Hall, J.; West, C.A.; Wisniewski, L.; Powers, T.W.; Angel, P.M.; Mehta, A.S.; Lewin, D.N.; Haab, B.B.; et al. Imaging Mass Spectrometry and Lectin Analysis of N-Linked Glycans in Carbohydrate Antigen-Defined Pancreatic Cancer Tissues. Mol. Cell Proteom. 2021, 20, 100012. [Google Scholar] [CrossRef]
  34. Da Fonseca, L.M.; Calvalhan, D.M.; Previato, J.O.; Mendonca Previato, L.; Freire-de-Lima, L. Resistance to paclitaxel induces glycophenotype changes and mesenchymal-to-epithelial transition activation in the human prostate cancer cell line PC-3. Tumour. Biol. 2020, 42, 1010428320957506. [Google Scholar] [CrossRef]
  35. Da Fonseca, L.M.; da Silva, V.A.; da Costa, K.M.; Dos Reis, J.S.; Previato, J.O.; Previato, L.M.; Freire-de-Lima, L. Resistance to cisplatin in human lung adenocarcinoma cells: Effects on the glycophenotype and epithelial to mesenchymal transition markers. Glycoconj J. 2022, 39, 247–259. [Google Scholar] [CrossRef]
  36. Ren, W.W.; Jin, Z.C.; Dong, W.; Kitajima, T.; Gao, X.D.; Fujita, M. Glycoengineering of HEK293 cells to produce high-mannose-type N-glycan structures. J. Biochem. 2019, 166, 245–258. [Google Scholar] [CrossRef]
  37. Boyaval, F.; Dalebout, H.; Van Zeijl, R.; Wang, W.; Farina-Sarasqueta, A.; Lageveen-Kammeijer, G.S.M.; Boonstra, J.J.; McDonnell, L.A.; Wuhrer, M.; Morreau, H.; et al. High-Mannose N-Glycans as Malignant Progression Markers in Early-Stage Colorectal Cancer. Cancers 2022, 14, 1552. [Google Scholar] [CrossRef]
  38. Sethi, M.K.; Hancock, W.S.; Fanayan, S. Identifying N-Glycan Biomarkers in Colorectal Cancer by Mass Spectrometry. Acc. Chem. Res. 2016, 49, 2099–2106. [Google Scholar] [CrossRef]
  39. Scupakova, K.; Adelaja, O.T.; Balluff, B.; Ayyappan, V.; Tressler, C.M.; Jenkinson, N.M.; Claes, B.S.; Bowman, A.P.; Cimino-Mathews, A.M.; White, M.J.; et al. Clinical importance of high-mannose, fucosylated, and complex N-glycans in breast cancer metastasis. JCI Insight 2021, 6, e146945. [Google Scholar] [CrossRef]
  40. Costa, A.F.; Campos, D.; Reis, C.A.; Gomes, C. Targeting Glycosylation: A New Road for Cancer Drug Discovery. Trends Cancer 2020, 6, 757–766. [Google Scholar] [CrossRef]
  41. Mockl, L. The Emerging Role of the Mammalian Glycocalyx in Functional Membrane Organization and Immune System Regulation. Front. Cell Dev. Biol. 2020, 8, 253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Ding, Y.; Gelfenbeyn, K.; Freire-de-Lima, L.; Handa, K.; Hakomori, S.I. Induction of epithelial-mesenchymal transition with O-glycosylated oncofetal fibronectin. FEBS Lett. 2012, 586, 1813–1820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Freire-de-Lima, L.; Gelfenbeyn, K.; Ding, Y.; Mandel, U.; Clausen, H.; Handa, K.; Hakomori, S.I. Involvement of O-glycosylation defining oncofetal fibronectin in epithelial-mesenchymal transition process. Proc. Natl. Acad. Sci. USA 2011, 108, 17690–17695. [Google Scholar] [CrossRef] [Green Version]
  44. Britain, C.M.; Bhalerao, N.; Silva, A.D.; Chakraborty, A.; Buchsbaum, D.J.; Crowley, M.R.; Crossman, D.K.; Edwards, Y.J.K.; Bellis, S.L. Glycosyltransferase ST6Gal-I promotes the epithelial to mesenchymal transition in pancreatic cancer cells. J. Biol. Chem. 2021, 296, 100034. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, J.; Ten Dijke, P.; Wuhrer, M.; Zhang, T. Role of glycosylation in TGF-beta signaling and epithelial-to-mesenchymal transition in cancer. Protein Cell 2021, 12, 89–106. [Google Scholar] [CrossRef]
  46. Pucci, M.; Malagolini, N.; Dall’Olio, F. Glycobiology of the Epithelial to Mesenchymal Transition. Biomedicines 2021, 9, 770. [Google Scholar] [CrossRef] [PubMed]
  47. Liao, C.; An, J.; Tan, Z.; Xu, F.; Liu, J.; Wang, Q. Changes in Protein Glycosylation in Head and Neck Squamous Cell Carcinoma. J. Cancer 2021, 12, 1455–1466. [Google Scholar] [CrossRef]
  48. Very, N.; Lefebvre, T.; El Yazidi-Belkoura, I. Drug resistance related to aberrant glycosylation in colorectal cancer. Oncotarget 2018, 9, 1380–1402. [Google Scholar] [CrossRef] [Green Version]
  49. Ferreira, J.A.; Peixoto, A.; Neves, M.; Gaiteiro, C.; Reis, C.A.; Assaraf, Y.G.; Santos, L.L. Mechanisms of cisplatin resistance and targeting of cancer stem cells: Adding glycosylation to the equation. Drug Resist. Updates 2016, 24, 34–54. [Google Scholar] [CrossRef] [Green Version]
  50. Wu, J.; Chen, S.; Liu, H.; Zhang, Z.; Ni, Z.; Chen, J.; Yang, Z.; Nie, Y.; Fan, D. Tunicamycin specifically aggravates ER stress and overcomes chemoresistance in multidrug-resistant gastric cancer cells by inhibiting N-glycosylation. J. Exp. Clin. Cancer Res. 2018, 37, 272. [Google Scholar] [CrossRef]
  51. Salgia, R.; Kulkarni, P. The Genetic/Non-genetic Duality of Drug ‘Resistance’ in Cancer. Trends Cancer 2018, 4, 110–118. [Google Scholar] [CrossRef] [PubMed]
  52. Abouelhadid, S.; Raynes, J.; Bui, T.; Cuccui, J.; Wren, B.W. Characterization of Posttranslationally Modified Multidrug Efflux Pumps Reveals an Unexpected Link between Glycosylation and Antimicrobial Resistance. mBio 2020, 11, e02604-20. [Google Scholar] [CrossRef] [PubMed]
  53. Kudo, T.; Nakagawa, H.; Takahashi, M.; Hamaguchi, J.; Kamiyama, N.; Yokoo, H.; Nakanishi, K.; Nakagawa, T.; Kamiyama, T.; Deguchi, K.; et al. N-glycan alterations are associated with drug resistance in human hepatocellular carcinoma. Mol. Cancer 2007, 6, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kizuka, Y.; Taniguchi, N. Enzymes for N-Glycan Branching and Their Genetic and Nongenetic Regulation in Cancer. Biomolecules 2016, 6, 25. [Google Scholar] [CrossRef] [Green Version]
  55. Assaraf, Y.G.; Brozovic, A.; Goncalves, A.C.; Jurkovicova, D.; Line, A.; Machuqueiro, M.; Saponara, S.; Sarmento-Ribeiro, A.B.; Xavier, C.P.R.; Vasconcelos, M.H. The multi-factorial nature of clinical multidrug resistance in cancer. Drug Resist. Updates 2019, 46, 100645. [Google Scholar] [CrossRef]
  56. Catalano, A.; Iacopetta, D.; Ceramella, J.; Scumaci, D.; Giuzio, F.; Saturnino, C.; Aquaro, S.; Rosano, C.; Sinicropi, M.S. Multidrug Resistance (MDR): A Widespread Phenomenon in Pharmacological Therapies. Molecules 2022, 27, 616. [Google Scholar] [CrossRef]
  57. Debnath, P.; Huirem, R.S.; Dutta, P.; Palchaudhuri, S. Epithelial-mesenchymal transition and its transcription factors. Biosci Rep. 2022, 42, BSR20211754. [Google Scholar] [CrossRef]
  58. Fedele, M.; Sgarra, R.; Battista, S.; Cerchia, L.; Manfioletti, G. The Epithelial-Mesenchymal Transition at the Crossroads between Metabolism and Tumor Progression. Int. J. Mol. Sci. 2022, 23, 800. [Google Scholar] [CrossRef]
  59. Qiao, L.; Chen, Y.; Liang, N.; Xie, J.; Deng, G.; Chen, F.; Wang, X.; Liu, F.; Li, Y.; Zhang, J. Targeting Epithelial-to-Mesenchymal Transition in Radioresistance: Crosslinked Mechanisms and Strategies. Front. Oncol. 2022, 12, 775238. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

dos Reis, J.S.; Rodrigues da Costa Santos, M.A.; Mendonça, D.P.; Martins do Nascimento, S.I.; Barcelos, P.M.; Correia de Lima, R.G.; da Costa, K.M.; Freire-de-Lima, C.G.; Morrot, A.; Previato, J.O.; et al. Glycobiology of Cancer: Sugar Drives the Show. Medicines 2022, 9, 34. https://0-doi-org.brum.beds.ac.uk/10.3390/medicines9060034

AMA Style

dos Reis JS, Rodrigues da Costa Santos MA, Mendonça DP, Martins do Nascimento SI, Barcelos PM, Correia de Lima RG, da Costa KM, Freire-de-Lima CG, Morrot A, Previato JO, et al. Glycobiology of Cancer: Sugar Drives the Show. Medicines. 2022; 9(6):34. https://0-doi-org.brum.beds.ac.uk/10.3390/medicines9060034

Chicago/Turabian Style

dos Reis, Jhenifer Santos, Marcos André Rodrigues da Costa Santos, Daniella Pereira Mendonça, Stefani Ingrid Martins do Nascimento, Pedro Marçal Barcelos, Rafaela Gomes Correia de Lima, Kelli Monteiro da Costa, Celio Geraldo Freire-de-Lima, Alexandre Morrot, Jose Osvaldo Previato, and et al. 2022. "Glycobiology of Cancer: Sugar Drives the Show" Medicines 9, no. 6: 34. https://0-doi-org.brum.beds.ac.uk/10.3390/medicines9060034

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop