Document Type : Review Article

Authors

1 Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China

2 Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran

Abstract

Protein is an important component of life. Protein modification after translation enriches the diversity of protein, regulates the structure and function of a protein, and participates in more life processes. Recent studies have found that post-translational modifications of proteins can regulate the occurrence and development of tumors. The human immune system should be able to eliminate cancer cells through an acquired immune response executed by T cells. However, clinical detection of cancer cells often results from the failure of immune surveillance. Therefore, relieving immune suppression and restoring antitumor immune response provides the possibility for tumor therapy. Tumor immunotherapy refers to exogenous intervention of the body's immune system, restart and maintain the "tumor-immune" cycle, restore and improve the anti-immune response of the group, strengthen the recognition and killing ability of tumor cells, so as to achieve the therapeutic effect of controlling or even clarifying the tumor specifically. Here, we review current knowledge of the current status of tumor immunotherapy and the types and effects of post-translational modifications of proteins, hoping to improve new ideas for the types of therapies.

Graphical Abstract

Post-translational modifications of proteins in tumor immunotherapy and their potential as immunotherapeutic targets

Keywords

Main Subjects

Selected author of this article by journal

ِDr. Mohammad Reza Mohammadi
Tarbiat Modares University

Google Scholar

Open Access

This article is licensed under a CC BY License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit: http://creativecommons.org/licenses/by/4.0/

 

Publisher’s Note

CMBR journal remains neutral with regard to jurisdictional claims in published maps and institutional afflictions.

 

Letters to Editor

Given that CMBR Journal's policy in accepting articles will be strict and will do its best to ensure that in addition to having the highest quality published articles, the published articles should have the least similarity (maximum 15%). Also, all the figures and tables in the article must be original and the copyright permission of images must be prepared by authors. However, some articles may have flaws and have passed the journal filter, which dear authors may find fault with. Therefore, the editor of the journal asks the authors, if they see an error in the published articles of the journal, to email the article information along with the documents to the journal office.

CMBR Journal welcomes letters to the editor ([email protected], [email protected]) for the post-publication discussions and corrections which allows debate post publication on its site, through the Letters to Editor. Critical letters can be sent to the journal editor as soon as the article is online. Following points are to be considering before sending the letters (comments) to the editor.

[1] Letters that include statements of statistics, facts, research, or theories should include appropriate references, although more than three are discouraged.

[2] Letters that are personal attacks on an author rather than thoughtful criticism of the author’s ideas will not be considered for publication.

[3] There is no limit to the number of words in a letter.

[4] Letter writers should include a statement at the beginning of the letter stating that it is being submitted either for publication or not.

[5] Anonymous letters will not be considered.

[6] Letter writers must include Name, Email Address, Affiliation, mobile phone number, and Comments.

[7] Letters will be answered as soon as possible.

  1. Zhang S-W, Wang H, Ding X-H, Xiao Y-L, Shao Z-M, You C, Gu Y-J, Jiang Y-Z (2022) Bidirectional crosstalk between therapeutic cancer vaccines and the tumor microenvironment: Beyond tumor antigens. Fundamental Research 2022: In Press. doi: https://doi.org/10.1016/j.fmre.2022.03.009
  2. Zhao H, Wu L, Yan G, Chen Y, Zhou M, Wu Y, Li Y (2021) Inflammation and tumor progression: signaling pathways and targeted intervention. Signal transduction and targeted therapy 6 (1): 263. doi: https://doi.org/10.1038/s41392-021-00658-5
  3. Whiteside TL Immune suppression in cancer: effects on immune cells, mechanisms and future therapeutic intervention. In: Seminars in cancer biology, 2006. vol 1. Elsevier, pp 3-15. doi: https://doi.org/10.1016/j.semcancer.2005.07.008
  4. Kennedy R, Celis E (2008) Multiple roles for CD4+ T cells in anti‐tumor immune responses. Immunological reviews 222 (1): 129-144. doi: https://doi.org/10.1111/j.1600-065X.2008.00616.x
  5. Xu Z, Zeng S, Gong Z, Yan Y (2020) Inmunoterapia basada en exosomas: un enfoque prometedor para el tratamiento del cáncer. Mol Cáncer 19 (1). doi: https://doi.org/10.1186/s12943-020-01278-3
  6. Melcher A, Harrington K, Vile R (2021) Oncolytic virotherapy as immunotherapy. Science 374 (6573): 1325-1326. doi: https://doi.org/10.1126/science.abk3436
  7. Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X (2022) Cancer vaccines as promising immuno-therapeutics: Platforms and current progress. Journal of Hematology & Oncology 15 (1): 28. doi: https://doi.org/10.1186/s13045-022-01247-x
  8. Parsonidis P, Papasotiriou I (2022) Adoptive Cellular Transfer Immunotherapies for Cancer. Cancer Treatment and Research Communications 32: 100575. doi: https://doi.org/10.1016/j.ctarc.2022.100575
  9. Lao Y, Shen D, Zhang W, He R, Jiang M (2022) Immune Checkpoint Inhibitors in Cancer Therapy—How to Overcome Drug Resistance? Cancers 14 (15): 3575. doi: https://doi.org/10.3390/cancers14153575
  10. de Miguel-Perez D, Russo A, Arrieta O, Ak M, Barron F, Gunasekaran M, Mamindla P, Lara-Mejia L, Peterson CB, Er ME, Peddagangireddy V, Buemi F, Cooper B, Manca P, Lapidus RG, Hsia R-C, Cardona AF, Naing A, Kaushal S, Hirsch FR, Mack PC, Serrano MJ, Adamo V, Colen RR, Rolfo C (2022) Extracellular vesicle PD-L1 dynamics predict durable response to immune-checkpoint inhibitors and survival in patients with non-small cell lung cancer. Journal of Experimental & Clinical Cancer Research 41 (1): 186. doi: https://doi.org/10.1186/s13046-022-02379-1
  11. Shevach EM, McHugh RS, Piccirillo CA, Thornton AM (2002) Control of T-cell activation by CD4+ CD25+ suppressor T cells. Immunological reviews 182 (1): 58-67. doi: https://doi.org/10.1034/j.1600-065X.2001.1820104.x
  12. Ramazi S, Zahiri J (2021) Post-translational modifications in proteins: resources, tools and prediction methods. Database 2021: baab012. doi: https://doi.org/10.1093/database/baab012
  13. Duan G, Walther D (2015) The Roles of Post-translational Modifications in the Context of Protein Interaction Networks. PLOS Computational Biology 11 (2): e1004049. doi: https://doi.org/10.1371/journal.pcbi.1004049
  14. Terman JR, Kashina A (2013) Post-translational modification and regulation of actin. Current Opinion in Cell Biology 25 (1): 30-38. doi: https://doi.org/10.1016/j.ceb.2012.10.009
  15. Deribe YL, Pawson T, Dikic I (2010) Post-translational modifications in signal integration. Nature Structural & Molecular Biology 17 (6): 666-672. doi: https://doi.org/10.1038/nsmb.1842
  16. Lei M-Z, Li X-X, Zhang Y, Li J-T, Zhang F, Wang Y-P, Yin M, Qu J, Lei Q-Y (2020) Acetylation promotes BCAT2 degradation to suppress BCAA catabolism and pancreatic cancer growth. Signal transduction and targeted therapy 5 (1): 70. doi: https://doi.org/10.1038/s41392-020-0168-0
  17. Zhu Y, Gu L, Lin X, Liu C, Lu B, Cui K, Zhou F, Zhao Q, Prochownik EV, Fan C, Li Y (2020) Dynamic Regulation of ME1 Phosphorylation and Acetylation Affects Lipid Metabolism and Colorectal Tumorigenesis. Molecular Cell 77 (1): 138-149.e135. doi: https://doi.org/10.1016/j.molcel.2019.10.015
  18. Zhao D, Zou S-W, Liu Y, Zhou X, Mo Y, Wang P, Xu Y-H, Dong B, Xiong Y, Lei Q-Y (2013) Lysine-5 acetylation negatively regulates lactate dehydrogenase A and is decreased in pancreatic cancer. Cancer cell 23 (4): 464-476. doi: https://doi.org/10.1016/j.ccr.2013.02.005
  19. Wang B, Ye Y, Yang X, Liu B, Wang Z, Chen S, Jiang K, Zhang W, Jiang H, Mustonen H (2020) SIRT 2‐dependent IDH 1 deacetylation inhibits colorectal cancer and liver metastases. EMBO reports 21 (4): e48183. doi: https://doi.org/10.15252/embr.201948183
  20. Cai Z, Li C-F, Han F, Liu C, Zhang A, Hsu C-C, Peng D, Zhang X, Jin G, Rezaeian A-H (2020) Phosphorylation of PDHA by AMPK drives TCA cycle to promote cancer metastasis. Molecular cell 80 (2): 263-278. e267. doi: https://doi.org/10.1016/j.molcel.2020.09.018
  21. Shao F, Yang X, Wang W, Wang J, Guo W, Feng X, Shi S, Xue Q, Gao S, Gao Y (2019) Associations of PGK1 promoter hypomethylation and PGK1‐mediated PDHK1 phosphorylation with cancer stage and prognosis: a TCGA pan‐cancer analysis. Cancer Communications 39 (1): 1-17. doi: https://doi.org/10.1186/s40880-019-0401-9
  22. Wang Y, Liu Q, Zhang H (2020) Phosphorylation of CREB-specific coactivator CRTC2 at Ser238 Promotes proliferation, migration, and invasion of colorectal cancer cells. Technology in Cancer Research & Treatment 19: 1533033820962111. doi: https://doi.org/10.1177/1533033820962111
  23. Eerola SK, Santio NM, Rinne S, Kouvonen P, Corthals GL, Scaravilli M, Scala G, Serra A, Greco D, Ruusuvuori P (2019) Phosphorylation of NFATC1 at PIM1 target sites is essential for its ability to promote prostate cancer cell migration and invasion. Cell Communication and Signaling 17 (1): 1-16. doi: https://doi.org/10.1186/s12964-019-0463-y
  24. Dall’Olio F, Malagolini N, Trinchera M, Chiricolo M (2012) Mechanisms of cancer-associated glycosylation changes. Frontiers in Bioscience-Landmark 17 (2): 670-699. doi:
  25. Jiang Y, Liu Z, Xu F, Dong X, Cheng Y, Hu Y, Gao T, Liu J, Yang L, Jia X (2018) Aberrant O‐glycosylation contributes to tumorigenesis in human colorectal cancer. Journal of cellular and molecular medicine 22 (10): 4875-4885. doi: https://doi.org/10.1111/jcmm.13752
  26. Löfgren C, Hjortsberg L, Blennow M, Lotfi K, Paul C, Eriksson S, Albertioni F (2004) Mechanisms of cross-resistance between nucleoside analogues and vincristine or daunorubicin in leukemic cells. Biochemical and biophysical research communications 320 (3): 825-832. doi: https://doi.org/10.1016/j.bbrc.2004.06.016
  27. Xiao L, Guan X, Xiang M, Wang Q, Long Q, Yue C, Chen L, Liu J, Liao C (2022) B7 family protein glycosylation: Promising novel targets in tumor treatment. Frontiers in Immunology 13: 1088560. doi: https://doi.org/10.3389%2Ffimmu.2022.1088560
  28. Yu Z, Jiang X, Qin L, Deng H, Wang J, Ren W, Li H, Zhao L, Liu H, Yan H (2021) A novel UBE2T inhibitor suppresses Wnt/β-catenin signaling hyperactivation and gastric cancer progression by blocking RACK1 ubiquitination. Oncogene 40 (5): 1027-1042. doi: https://doi.org/10.1038/s41388-020-01572-w
  29. Ciechanover A (2005) Intracellular protein degradation: from a vague idea thru the lysosome and the ubiquitin–proteasome system and onto human diseases and drug targeting. Cell Death & Differentiation 12 (9): 1178-1190. doi: https://doi.org/10.1038/sj.cdd.4401692
  30. Li W, Shen M, Jiang Y-Z, Zhang R, Zheng H, Wei Y, Shao Z-M, Kang Y (2020) Deubiquitinase USP20 promotes breast cancer metastasis by stabilizing SNAI2. Genes & development 34 (19-20): 1310-1315. doi: https://doi.org/10.1101/gad.339804.120
  31. Colegio OR, Chu N-Q, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM (2014) Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513 (7519): 559-563. doi: https://doi.org/10.1038/nature13490
  32. Dai M, Wang L, Yang J, Chen J, Dou X, Chen R, Ge Y, Lin Y (2023) LDHA as a regulator of T cell fate and its mechanisms in disease. Biomedicine & Pharmacotherapy 158: 114164. doi: https://doi.org/10.1016/j.biopha.2022.114164
  33. Javaid N, Choi S (2017) Acetylation-and methylation-related epigenetic proteins in the context of their targets. Genes 8 (8): 196. doi: https://doi.org/10.3390/genes8080196
  34. Wan J, Liu H, Ming L (2019) Lysine crotonylation is involved in hepatocellular carcinoma progression. Biomedicine & Pharmacotherapy 111: 976-982. doi: https://doi.org/10.1016/j.biopha.2018.12.148
  35. Guzy RD, Sharma B, Bell E, Chandel NS, Schumacker PT (2008) Loss of the SdhB, but Not the SdhA, subunit of complex II triggers reactive oxygen species-dependent hypoxia-inducible factor activation and tumorigenesis. Molecular and cellular biology 28 (2): 718-731. doi: https://doi.org/10.1128/MCB.01338-07
  36. Ren M, Yang X, Bie J, Wang Z, Liu M, Li Y, Shao G, Luo J (2020) Citrate synthase desuccinylation by SIRT5 promotes colon cancer cell proliferation and migration. Biological Chemistry 401 (9): 1031-1039. doi: https://doi.org/10.1515/hsz-2020-0118
  37. Colak G, Pougovkina O, Dai L, Tan M, Te Brinke H, Huang H, Cheng Z, Park J, Wan X, Liu X (2015) Proteomic and Biochemical Studies of Lysine Malonylation Suggest Its Malonic Aciduria-associated Regulatory Role in Mitochondrial Function and Fatty Acid Oxidation [S]. Molecular & Cellular Proteomics 14 (11): 3056-3071. doi: https://doi.org/10.1074/mcp.M115.048850
  38. Bruning U, Morales-Rodriguez F, Kalucka J, Goveia J, Taverna F, Queiroz KC, Dubois C, Cantelmo AR, Chen R, Loroch S (2018) Impairment of angiogenesis by fatty acid synthase inhibition involves mTOR malonylation. Cell metabolism 28 (6): 866-880. e815. doi: https://doi.org/10.1016/j.cmet.2018.07.019
  39. Huang Q, Wu D, Zhao J, Yan Z, Chen L, Guo S, Wang D, Yuan C, Wang Y, Liu X (2022) TFAM loss induces nuclear actin assembly upon mDia2 malonylation to promote liver cancer metastasis. The EMBO journal 41 (11): e110324. doi: https://doi.org/10.15252/embj.2021110324
  40. Munnur D, Bartlett E, Mikolčević P, Kirby IT, Rack JGM, Mikoč A, Cohen MS, Ahel I (2019) Reversible ADP-ribosylation of RNA. Nucleic acids research 47 (11): 5658-5669. doi: https://doi.org/10.1093/nar/gkz305
  41. Li W, Li F, Zhang X, Lin H-K, Xu C (2021) Insights into the post-translational modification and its emerging role in shaping the tumor microenvironment. Signal Transduction and Targeted Therapy 6 (1): 422. doi: https://doi.org/10.1038/s41392-021-00825-8
  42. Yu X, Li W, Young KH, Li Y (2021) Posttranslational modifications in PD-L1 turnover and function: from cradle to Grave. Biomedicines 9 (11): 1702. doi: https://doi.org/10.3390/biomedicines9111702
  43. Shi C, Wang Y, Wu M, Chen Y, Liu F, Shen Z, Wang Y, Xie S, Shen Y, Sang L (2022) Promoting anti-tumor immunity by targeting TMUB1 to modulate PD-L1 polyubiquitination and glycosylation. Nature Communications 13 (1): 6951. doi: https://doi.org/10.1038/s41467-022-34346-x
  44. Kao C-F, Chen S-Y, Chen J-Y, Wu Lee Y-H (2004) Modulation of p53 transcription regulatory activity and post-translational modification by hepatitis C virus core protein. Oncogene 23 (14): 2472-2483. doi: https://doi.org/10.1038/sj.onc.1207368
  45. Li C, Zhang S, Qiu T, Wang Y, Ricketts DM, Qi C (2019) Upregulation of long non-coding RNA NNT-AS1 promotes osteosarcoma progression by inhibiting the tumor suppressive miR-320a. Cancer biology & therapy 20 (4): 413-422. doi: https://doi.org/10.1080/15384101.2017.1346760
  46. Cohen HY, Lavu S, Bitterman KJ, Hekking B, Imahiyerobo TA, Miller C, Frye R, Ploegh H, Kessler BM, Sinclair DA (2004) Acetylation of the C terminus of Ku70 by CBP and PCAF controls Bax-mediated apoptosis. Molecular cell 13 (5): 627-638. doi: https://doi.org/10.1016/S1097-2765(04)00094-2
  47. Yang Y, Ye W-L, Zhang R-N, He X-S, Wang J-R, Liu Y-X, Wang Y, Yang X-M, Zhang Y-J, Gan W-J (2021) The role of TGF-β signaling pathways in cancer and its potential as a therapeutic target. Evidence-Based Complementary and Alternative Medicine 2021: Article ID 6675208. doi: https://doi.org/10.1155/2021/6675208
  48. Lynch SJ, Snitkin H, Gumper I, Philips MR, Sabatini D, Pellicer A (2015) The differential palmitoylation states of N‐Ras and H‐Ras determine their distinct Golgi subcompartment localizations. Journal of cellular physiology 230 (3): 610-619. doi: https://doi.org/10.1002/jcp.24779
  49. Sung PJ, Tsai FD, Vais H (2013) Phosphorylated K-Ras limits cell survival by blocking Bcl-xL sensitization of inositol trisphosphate receptors. Proc Natl Acad Sci USA 110 (51): 20593-20598. doi: https://doi.org/10.1073/pnas.1306431110
  50. Jura N, Scotto-Lavino E, Sobczyk A, Bar-Sagi D (2006) Differential modification of Ras proteins by ubiquitination. Molecular cell 21 (5): 679-687. doi: https://doi.org/10.1016/j.molcel.2006.02.011
  51. Brantley EC, Nabors LB, Gillespie GY, Choi Y-H, Palmer CA, Harrison K, Roarty K, Benveniste EN (2008) Loss of protein inhibitors of activated STAT-3 expression in glioblastoma multiforme tumors: implications for STAT-3 activation and gene expression. Clinical cancer research 14 (15): 4694-4704. doi: https://doi.org/10.1158/1078-0432.CCR-08-0618
  52. Bradley CA (2018) Efficacy of a PSMA-targeted nanoparticle. Nature Reviews Urology 15 (10): 590-591. doi: https://doi.org/10.1038/s41585-018-0067-0
  53. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH (2013) The somatic genomic landscape of glioblastoma. cell 155 (2): 462-477. doi: https://doi.org/10.1016/j.cell.2013.09.034
  54. Recio C, Guerra B, Guerra-Rodriguez M, Aranda-Tavio H, Martín-Rodríguez P, de Mirecki-Garrido M, Brito-Casillas Y, García-Castellano JM, Estévez-Braun A, Fernández-Pérez L (2019) Signal transducer and activator of transcription (STAT)-5: An opportunity for drug development in oncohematology. Oncogene 38 (24): 4657-4668. doi: https://doi.org/10.1038/s41388-019-0752-3
  55. Lalle G, Twardowski J, Grinberg-Bleyer Y (2021) NF-κB in cancer immunity: friend or foe? Cells 10 (2): 355. doi: https://doi.org/10.3390/cells10020355
  56. Imada K, Leonard WJ (2000) The jak-STAT pathway. Molecular immunology 37 (1-2): 1-11. doi: https://doi.org/10.1016/S0161-5890(00)00018-3
  57. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A (2015) Tissue-based map of the human proteome. Science 347 (6220): 1260419. doi: https://doi.org/10.1126/science.1260419
  58. Feng HB, Chen Y, Xie Z, Jiang J, Zhong YM, Guo WB, Yan WQ, Lv ZY, Lu DX, Liang HL (2021) High SHP2 expression determines the efficacy of PD‐1/PD‐L1 inhibitors in advanced KRAS mutant non‐small cell lung cancer. Thoracic Cancer 12 (19): 2564-2573. doi: https://doi.org/10.1111/1759-7714.14137
  59. Tainsky MA (2009) Genomic and proteomic biomarkers for cancer: a multitude of opportunities. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer 1796 (2): 176-193. doi: https://doi.org/10.1016/j.bbcan.2009.04.004