Skip to content
2000
Volume 25, Issue 15
  • ISSN: 1871-5303
  • E-ISSN: 2212-3873

Abstract

Aim

The aim of this study is to examine the role of the microrchidia (MORC) family, a group of chromatin remodeling proteins, as the therapeutic and prognostic markers for colorectal cancer (CRC).

Background

MORC protein family genes are a highly conserved nucleoprotein superfamily whose members share a common domain but have distinct biological functions. Previous studies have analyzed the roles of MORCs as epigenetic regulators and chromatin remodulators; however, the involvement of MORCs in the development and pathogenesis of CRC was less examined.

Objective

The current work examined the role of the MORCs as the therapeutic and prognostic markers for CRC.

Methods

The expressions and prognostic significance of MORC family genes in CRC were explored. The role of these genes in tumor immunity was comprehensively analyzed in terms of their functions in immune cell infiltration, tumor microenvironment (TME), and their interaction with immune regulatory genes such as immunosuppressive genes, immune checkpoints and immunostimulatory genes. The relations between MORC family genes, tumor mutation burden (TMB), DNA, mismatch repair (MMR), RNA methylation, microsatellite instability (MSI), and drug sensitivity were investigated using the R statistical software. The expressions of MORC4 in 150 CRC tissues and 60 paracancer tissues were detected by immunohistochemical method. CRC cell proliferation, migration, and invasion were measured by cell counting kit-8 (CCK-8), scratch assay, and transwell cell invasion assay.

Results

The expressions of MORC2 and MORC4 were significantly upregulated, whereas those of MORC1 and MORC3 were noticeably downregulated in CRC in comparison to their expressions in normal colorectal mucosal tissues. Patients with high-expressed MORC2 showed a more unfavorable prognosis than those with a low MORC2 level. Functional annotation analysis identified 100 MORC family genes with the most significant negative or positive correlations to diabetic cardiomyopathy, amyotrophic lateral sclerosis, oxidative phosphorylation, Huntington’s disease, thermogenesis, Parkinson’s disease, olfactory transduction, Alzheimer’s disease, prion disease. MORC3 expression was positively correlated with Stromal score, Immune score and ESTIMATE score, while MORC2 expression was negatively related to the three scores in CRC, these correlations were not statistically significant. Additionally, the MORC family genes were significantly positively correlated with tumor-infiltrating immune cells such as T helper cells and exhibited close relations to some immunosuppressive genes such as CXCR4 and PVR, immunostimulatory genes such as TGFBR1, KDR, and CD160 as well as some immune checkpoint genes. It was found that the expressions of some members of MORC family genes were positively correlated with DNA methylation, MSI, TMB, MMRs, and drug sensitivity in CRC and that the mRNA and protein levels of MORC4 were remarkably upregulated in CRC tissues than in adjacent normal tissues (<0.05). In the MORC4 knockdown group, DLD-1 cell proliferation was more inhibited than in the negative control (NC) and siRNA groups (<0.05). Furthermore, the migratory capacity of DLD-1 cells and the number of cells crossing the basement membrane in the MORC4 knockdown group were reduced compared to the NC and siRNA groups (all <0.05).

Conclusion

The expressions of MORC family genes were significantly different in CRC samples, which was related to the immune cell infiltration and prognosis of CRC. Thus, the MORC family genes were considered as markers for indicating the clinical immunotherapy and prognostic outcome of CRC.

This is an open access article published under CC BY 4.0 https://creativecommons.org/licenses/by/4.0/legalcode
Loading

Article metrics loading...

/content/journals/emiddt/10.2174/0118715303367767241231113110
2025-01-13
2025-12-20
Loading full text...

Full text loading...

/deliver/fulltext/emiddt/25/15/EMIDDT-25-15-03.html?itemId=/content/journals/emiddt/10.2174/0118715303367767241231113110&mimeType=html&fmt=ahah

References

  1. ChangJ. LinG. YeM. TongD. ZhaoJ. ZhuD. YuQ. ZhangW. LiW. Decreased mean platelet volume predicts poor prognosis in metastatic colorectal cancer patients treated with first-line chemotherapy: Results from mCRC biomarker study.BMC Cancer20191911510.1186/s12885‑018‑5252‑230612568
    [Google Scholar]
  2. SunH. WangH. LiX. HaoY. LingJ. WangH. WangF. XuF. Increased MAD2L2 expression predicts poor clinical outcome in colon adenocarcinoma.Biocell202347360761810.32604/biocell.2023.026445
    [Google Scholar]
  3. NematiM. RasmiY. RezaieJ. Therapeutic application of mesenchymal stem cells-derived extracellular vesicles in colorectal cancer.Biocell202347345546410.32604/biocell.2023.025603
    [Google Scholar]
  4. ArnoldM. SierraM.S. LaversanneM. SoerjomataramI. JemalA. BrayF. Global patterns and trends in colorectal cancer incidence and mortality.Gut201766468369110.1136/gutjnl‑2015‑31091226818619
    [Google Scholar]
  5. RuanY. LuG. YuY. LuoY. WuH. ShenY. GaoZ. ShenY. CaiZ. LiL. PF-04449913 inhibits proliferation and metastasis of colorectal cancer cells by down-regulating MMP9 expression through the ERK/p65 pathway.Curr. Mol. Pharmacol.2024171e15092322116437724680
    [Google Scholar]
  6. ManoharS.M. JoshiK.S. Promising anticancer activity of multitarget cyclin dependent kinase inhibitors against human colorectal carcinoma cells.Curr. Mol. Pharmacol.20221571024103310.2174/187446721566622012412580935068399
    [Google Scholar]
  7. ChenW. ZhengR. BaadeP.D. ZhangS. ZengH. BrayF. JemalA. YuX.Q. HeJ. Cancer statistics in China, 2015.CA Cancer J. Clin.201666211513210.3322/caac.2133826808342
    [Google Scholar]
  8. WangH. ZhangL. LuoQ. LiuJ. WangG. MORC protein family-related signature within human disease and cancer.Cell Death Dis.20211212111210.1038/s41419‑021‑04393‑134839357
    [Google Scholar]
  9. ZhangS. GuoA. WangH. LiuJ. DongC. RenJ. WangG. Oncogenic MORC2 in cancer development and beyond.Genes Dis.202411286187310.1016/j.gendis.2023.05.01037692502
    [Google Scholar]
  10. KimH. YenL. WongpaleeS.P. KirshnerJ.A. MehtaN. XueY. JohnstonJ.B. BurlingameA.L. KimJ.K. LoparoJ.J. JacobsenS.E. The gene-silencing protein MORC-1 topologically entraps DNA and forms multimeric assemblies to cause DNA compaction.Mol. Cell2019754700710.e610.1016/j.molcel.2019.07.03231442422
    [Google Scholar]
  11. LiD.Q. NairS.S. KumarR. The MORC family.Epigenetics20138768569310.4161/epi.2497623804034
    [Google Scholar]
  12. PanZ. DingQ. GuoQ. GuoY. WuL. WuL. TangM. YuH. ZhouF. MORC2, a novel oncogene, is upregulated in liver cancer and contributes to proliferation, metastasis and chemoresistance.Int. J. Oncol.2018531597210.3892/ijo.2018.433329620211
    [Google Scholar]
  13. YangZ. ZhuangQ. HuG. GengS. MORC4 is a novel breast cancer oncogene regulated by miR-193b-3p.J. Cell. Biochem.201912034634464310.1002/jcb.2775130320920
    [Google Scholar]
  14. LiuM. SunX. ShiS. MORC2 enhances tumor growth by promoting angiogenesis and tumor-associated macrophage recruitment via Wnt/β-catenin in lung cancer.Cell. Physiol. Biochem.: Int. J. Experi. Cell. Physiol., Biochem., Pharmacol.20185141679169410.1159/00049567330504718
    [Google Scholar]
  15. SongZ. YuJ. WangM. ShenW. WangC. LuT. ShanG. DongG. WangY. ZhaoJ. CHDTEPDB: Transcriptome expression profile database and interactive analysis platform for congenital heart disease.Congenit. Heart Dis.202318669370110.32604/chd.2024.048081
    [Google Scholar]
  16. LiuW. YeH. LiuY.F. XuC.Q. ZhongY.X. TianT. MaS.W. TaoH. LiL. XueL.C. HeH.Q. Transcriptome-derived stromal and immune scores infer clinical outcomes of patients with cancer.Oncol. Lett.20181544351435710.3892/ol.2018.785529541203
    [Google Scholar]
  17. MarabelleA. FakihM. LopezJ. ShahM. Shapira-FrommerR. NakagawaK. ChungH.C. KindlerH.L. Lopez-MartinJ.A. MillerW.H.Jr ItalianoA. KaoS. Piha-PaulS.A. DelordJ.P. McWilliamsR.R. FabrizioD.A. Aurora-GargD. XuL. JinF. NorwoodK. BangY.J. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study.Lancet Oncol.202021101353136510.1016/S1470‑2045(20)30445‑932919526
    [Google Scholar]
  18. NishiyamaA. NakanishiM. Navigating the DNA methylation landscape of cancer.Trends Genet.202137111012102710.1016/j.tig.2021.05.00234120771
    [Google Scholar]
  19. YuanH. YanM. ZhangG. LiuW. DengC. LiaoG. XuL. LuoT. YanH. LongZ. ShiA. ZhaoT. XiaoY. LiX. CancerS.E.A. CancerSEA: A cancer single-cell state atlas.Nucleic Acids Res.201947D1D900D90810.1093/nar/gky93930329142
    [Google Scholar]
  20. LiuC.J. HuF.F. XieG.Y. MiaoY.R. LiX.W. ZengY. GuoA.Y. GSCA: An integrated platform for gene set cancer analysis at genomic, pharmacogenomic and immunogenomic levels.Brief. Bioinform.2023241bbac55810.1093/bib/bbac55836549921
    [Google Scholar]
  21. ZhangZ. PanY. ZhaoY. RenM. LiY. FengY. LuG. HeS. Knockdown Wiskott-Aldrich syndrome protein family member 3 (WASF3) inhibits colorectal cancer metastasis and sensitizes to cisplatin through targeting ZNF471.Biocell20224681917192410.32604/biocell.2022.018847
    [Google Scholar]
  22. SiegelR.L. MillerK.D. Goding SauerA. FedewaS.A. ButterlyL.F. AndersonJ.C. CercekA. SmithR.A. JemalA. Colorectal cancer statistics, 2020.CA Cancer J. Clin.202070314516410.3322/caac.2160132133645
    [Google Scholar]
  23. TakakuraY. HinoiT. EgiH. ShimomuraM. AdachiT. SaitoY. TanimineN. MiguchiM. OhdanH. Procalcitonin as a predictive marker for surgical site infection in elective colorectal cancer surgery.Langenbecks Arch. Surg.2013398683383910.1007/s00423‑013‑1095‑023784676
    [Google Scholar]
  24. MulitaF. LiolisE. AkinosoglouK. TchabashviliL. MaroulisI. KaplanisC. VailasM. PanosG. Postoperative sepsis after colorectal surgery: A prospective single-center observational study and review of the literature.Prz. Gastroenterol.2022171475110.5114/pg.2021.10608335371356
    [Google Scholar]
  25. HuhJ.W. LeeW.Y. ParkY.A. ChoY.B. KimH.C. YunS.H. ChunH.K. Oncological outcome of surgical site infection after colorectal cancer surgery.Int. J. Colorectal Dis.201934227728310.1007/s00384‑018‑3194‑430426197
    [Google Scholar]
  26. WangY. LiX. YuY. LiangJ. Risk factors for sepsis in patients with colorectal cancer complicated with gastrointestinal perforation and its impact on prognosis.J. Gastrointest. Oncol.202314280681410.21037/jgo‑23‑20537201047
    [Google Scholar]
  27. KlockerE.V. BarthD.A. RiedlJ.M. PrinzF. SzkanderaJ. SchlickK. KornpratP. LacknerK. LindenmannJ. StögerH. StotzM. GergerA. PichlerM. Decreased activity of circulating butyrylcholinesterase in blood is an independent prognostic marker in pancreatic cancer patients.Cancers (Basel)2020125115410.3390/cancers1205115432375339
    [Google Scholar]
  28. DimopoulosM.P. VerrasG.I. MulitaF. Editorial: Newest challenges and advances in the treatment of colorectal disorders; from predictive biomarkers to minimally invasive techniques.Front. Surg.202411148787810.3389/fsurg.2024.148787839469539
    [Google Scholar]
  29. CoulterD.W. BoettnerA.D. KortylewiczZ.P. EnkeS.P. LutherJ.A. VermaV. Baranowska-KortylewiczJ. Butyrylcholinesterase as a blood biomarker in neuroblastoma.J. Pediatr. Hematol. Oncol.201739427228110.1097/MPH.000000000000082828375942
    [Google Scholar]
  30. WangS. HuangX. ZhaoS. LvJ. LiY. WangS. GuoJ. WangY. WangR. ZhangM. QiuW. Progressions of the correlation between lipid metabolism and immune infiltration characteristics in gastric cancer and identification of BCHE as a potential biomarker.Front. Immunol.202415132756510.3389/fimmu.2024.132756538357546
    [Google Scholar]
  31. PerryJ. ZhaoY. The CW domain, a structural module shared amongst vertebrates, vertebrate-infecting parasites and higher plants.Trends Biochem. Sci.2003281157658010.1016/j.tibs.2003.09.00714607086
    [Google Scholar]
  32. LiangY. WuD. QuQ. LiZ. YinH. MORC4 plays a tumor-promoting role in colorectal cancer via regulating PCGF1/CDKN1A axis in vitro and in vivo.Cancer Gene Ther.202330798599610.1038/s41417‑023‑00605‑236932196
    [Google Scholar]
  33. ZhaoP. NingJ. HuangJ. WeiB. WangZ. HuangX. High expression of MORC2 is associated with poor clinical outcomes and immune infiltrates in colon adenocarcinoma.Int. J. Gen. Med.2023164595461510.2147/IJGM.S42071537850194
    [Google Scholar]
  34. YiF.S. ZhaiK. ShiH.Z. Helper T cells in malignant pleural effusion.Cancer Lett.2021500212810.1016/j.canlet.2020.12.01633309856
    [Google Scholar]
  35. MengQ. ZhaoY. XuM. WangP. LiJ. CuiR. FuW. DingS. Increased circulating regulatory T cells and decreased follicular T helper cells are associated with colorectal carcinogenesis.Front. Immunol.202415128763210.3389/fimmu.2024.128763238343544
    [Google Scholar]
  36. McGranahanN. FurnessA.J.S. RosenthalR. RamskovS. LyngaaR. SainiS.K. Jamal-HanjaniM. WilsonG.A. BirkbakN.J. HileyC.T. WatkinsT.B.K. ShafiS. MurugaesuN. MitterR. AkarcaA.U. LinaresJ. MarafiotiT. HenryJ.Y. Van AllenE.M. MiaoD. SchillingB. SchadendorfD. GarrawayL.A. MakarovV. RizviN.A. SnyderA. HellmannM.D. MerghoubT. WolchokJ.D. ShuklaS.A. WuC.J. PeggsK.S. ChanT.A. HadrupS.R. QuezadaS.A. SwantonC. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade.Science201635162801463146910.1126/science.aaf149026940869
    [Google Scholar]
  37. MarabelleA. LeD.T. AsciertoP.A. Di GiacomoA.M. De Jesus-AcostaA. DelordJ.P. GevaR. GottfriedM. PenelN. HansenA.R. Piha-PaulS.A. DoiT. GaoB. ChungH.C. Lopez-MartinJ. BangY.J. FrommerR.S. ShahM. GhoriR. JoeA.K. PruittS.K. DiazL.A.Jr Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: Results from the phase II KEYNOTE-158 study.J. Clin. Oncol.202038111010.1200/JCO.19.0210531682550
    [Google Scholar]
  38. JonesP.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond.Nat. Rev. Genet.201213748449210.1038/nrg323022641018
    [Google Scholar]
  39. DeatonA.M. BirdA. CpG islands and the regulation of transcription.Genes Dev.201125101010102210.1101/gad.203751121576262
    [Google Scholar]
  40. MundorfA. SchmitzJ. GüntürkünO. FreundN. OcklenburgS. Methylation of MORC1: A possible biomarker for depression?J. Psychiatr. Res.201810320821110.1016/j.jpsychires.2018.05.02629890506
    [Google Scholar]
  41. WangT. QinZ. WenL. GuoY. LiuQ. LeiZ. PanW. LiuK. WangX. LaiS. SunW. WeiY. LiuL. GuoL. ChenY. WangJ. XiaoH. BianX. ChenD. WangB. Epigenetic restriction of Hippo signaling by MORC2 underlies stemness of hepatocellular carcinoma cells.Cell Death Differ.201825122086210010.1038/s41418‑018‑0095‑629555977
    [Google Scholar]
  42. DuanX. GuoG. PeiX. WangX. LiL. XiongY. QiuX. Baicalin inhibits cell viability, migration and invasion in breast cancer by regulating miR-338-3p and MORC4.OncoTargets Ther.201912111831119310.2147/OTT.S21710131908485
    [Google Scholar]
/content/journals/emiddt/10.2174/0118715303367767241231113110
Loading
/content/journals/emiddt/10.2174/0118715303367767241231113110
Loading

Data & Media loading...

Supplements

Supplementary material is available on the publisher's website along with the published article.


  • Article Type:
    Research Article
Keyword(s): cell invasion; CRC; MORC4; prognosis; TCGA; tumor immunity
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test