Journal Home > Volume 28 , Issue 4

Gastric cancer (GC) is one of the most common cancers and ranks the third in cancer mortality all over the world. The goal of this study was to identify potential hub-genes, highlighting their functions, signaling pathways, and candidate drugs for the treatment of GC patients. We used publicly available next generation sequencing (NGS) data to identify differentially expressed (DE) genes. The top DE genes were mapped to STRING database to construct the protein-protein interaction (PPI) network and top hub genes were selected for further analysis. We found a total of 1555 DE genes with 870 upregulated and 685 downregulated genes in GC. We selected the top 400 (200 upregulated and 200 downregulated) genes to construct a PPI network and extracted the top 15 hub genes. The gene ontology (GO) term and kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses of the 15 hub genes exposed some important functions and signaling pathways that were significantly associated with GC patients. The survival analysis of the hub genes disclosed that the lower expressions of the three hub genes CDH2, COL4A1, and COL5A2 were associated with better survival of GC patients. These three genes might be the candidate biomarkers for the diagnosis and treatment of GC. Then, we considered 3 key proteins (genomic biomarkers) (COL4A1, CDH2, and CO5A2) as the drug target proteins (receptors), performed their docking analysis with the 102 meta-drug agents, and found Everolimus, Docetaxel, Lanreotide, Venetoclax, Temsirolimus, and Nilotinib as the top ranked 6 candidate drugs with respect to our proposed target proteins for the treatment against GC patients. Therefore, the proposed drugs might play vital role for the treatment against GC patients.


menu
Abstract
Full text
Outline
Electronic supplementary material
About this article

Identification of Key Genes as Potential Drug Targets for Gastric Cancer

Show Author's information Md. Tofazzal Hossain1,2,3,Md. Selim Reza1,2,Yin Peng4Shengzhong Feng1Yanjie Wei1( )
Center for High Performance Computing, Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
School of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
Guangdong Provincial Key Laboratory for Genome Stability and Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University, Shenzhen 518060, China

Md. Tofazzal Hossain and Md. Selim Reza contribute equally to this paper.

Abstract

Gastric cancer (GC) is one of the most common cancers and ranks the third in cancer mortality all over the world. The goal of this study was to identify potential hub-genes, highlighting their functions, signaling pathways, and candidate drugs for the treatment of GC patients. We used publicly available next generation sequencing (NGS) data to identify differentially expressed (DE) genes. The top DE genes were mapped to STRING database to construct the protein-protein interaction (PPI) network and top hub genes were selected for further analysis. We found a total of 1555 DE genes with 870 upregulated and 685 downregulated genes in GC. We selected the top 400 (200 upregulated and 200 downregulated) genes to construct a PPI network and extracted the top 15 hub genes. The gene ontology (GO) term and kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses of the 15 hub genes exposed some important functions and signaling pathways that were significantly associated with GC patients. The survival analysis of the hub genes disclosed that the lower expressions of the three hub genes CDH2, COL4A1, and COL5A2 were associated with better survival of GC patients. These three genes might be the candidate biomarkers for the diagnosis and treatment of GC. Then, we considered 3 key proteins (genomic biomarkers) (COL4A1, CDH2, and CO5A2) as the drug target proteins (receptors), performed their docking analysis with the 102 meta-drug agents, and found Everolimus, Docetaxel, Lanreotide, Venetoclax, Temsirolimus, and Nilotinib as the top ranked 6 candidate drugs with respect to our proposed target proteins for the treatment against GC patients. Therefore, the proposed drugs might play vital role for the treatment against GC patients.

Keywords: molecular docking, gastric cancer, hub genes, candidate genes, candidate drugs

References(80)

[1]
B. Freddie, F. Jacques, S. Isabelle, R. L. Siegel, L. A. Torre, and J. Ahmedin, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA. Cancer J. Clin., vol. 68, no. 6, pp. 394–424, 2018.
[2]
M. Rugge, R. M. Genta, F. D. Mario, E. M. El-Omar, H. B. El-Serag, M. Fassan, R. H. Hunt, E. J. Kuipers, P. Malfertheiner, K. Sugano, et al., Gastric cancer as preventable disease, Clin. Gastroenterol. Hepatol., .
[3]
Y. Shi, L. Qi, H. Chen, J. Zhang, Q. Guan, J. He, M. Li, Z. Guo, H. Yan, and P. Li, Identification of genes universally differentially expressed in gastric cancer, Biomed Res. Int., .
[4]
E. C. Smyth, M. Nilsson, H. I. Grabsch, N. C. V. Grieken, and F. Lordick, Gastric cancer, The Lancet, .
[5]
J. Machlowska, J. Baj, M. Sitarz, R. Maciejewski, and R. Sitarz, Gastric cancer: Epidemiology, risk factors, classification, genomic characteristics and treatment strategies, International Journal of Molecular Sciences, .
[6]
A. R. Yusefi, K. B. Lankarani, P. Bastani, M. Radinmanesh, and Z. Kavosi, Risk factors for gastric cancer: A systematic review, Asian Pacific Journal of Cancer Prevention, .
[7]
G. Herrera-Almario and V. E. Strong, Minimally invasive gastric surgery, Ann. Surg. Oncol., .
[8]
J. Tey, Y. Y. Soon, W. Y. Koh, C. N. Leong, B. Vellayappan, K. Lim, and I. W. K. Ivan, Palliative radiotherapy for gastric cancer: A systematic review and meta-analysis, Oncotarget, .
[9]
O. Zhandossov, G. Kaussova, and A. Koten, Combined treatment for gastric cancer: Immunological approach, Turkish J. Gastroenterol., .
[10]
E. Niccolai, A. Taddei, D. Prisco, and A. Amedei, Gastric cancer and the epoch of immunotherapy approaches, World J. Gastroenterol., .
[11]
L. Necula, L. Matei, D. Dragu, A. I. Neagu, C. Mambet, S. Nedeianu, C. Bleotu, C. C. Diaconu, and M. Chivu-Economescu, Recent advances in gastric cancer early diagnosis, World J. Gastroenterol., .
[12]
A. S. Crystal, A. T. Shaw, L. V. Sequist, L. Friboulet, M. J. Niederst, E. L. Lockerman, R. L. Frias, J. F. Gainor, A. Amzallag, P. Greninger, et al., Patient-derived models of acquired resistance can identify effective drug combinations for cancer, Science, .
[13]
A. A. Borisy, P. J. Elliott, N. W. Hurst, M. S. Lee, J. Lehar, E. R. Price, G. Serbedzija, G. R. Zimmermann, M. A. Foley, B. R. Stockwell, et al., Systematic discovery of multicomponent therapeutics, Proc. Natl. Acad. Sci., .
[14]
B. Klinger, A. Sieber, R. Fritsche-Guenther, F. Witzel, L. Berry, D. Schumacher, Y. Yan, P. Durek, M. Merchant, R. Schafer, et al., Network quantification of EGFR signaling unveils potential for targeted combination therapy, Mol. Syst. Biol., .
[15]
M. Bansal, J. Yang, C. Karan, M. P. Menden, J. C. Costello, H. Tang, G. Xiao, Y. Li, J. Allen, R. Zhong, et al., A community computational challenge to predict the activity of pairs of compounds, Nat. Biotechnol., .
[16]
L. A. M. Griner, R. Guha, P. Shinn, R. M. Young, J. M. Keller, D. Liu, I. S. Goldlust, A. Yasgar, C. MaKnight, M. B. Boxer, et al., High-throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell-like diffuse large B-cell lymphoma cells, Proc. Natl. Acad. Sci., .
[17]
M. L. Miller, E. J. Molinelli, J. S. Nair, T. Sheikh, R. Samy, X. Jing, Q. He, A. Korkut, A. M. Crago, S. Singer, et al., Drug synergy screen and network modeling in dedifferentiated liposarcoma identifies CDK4 and IGF1R as synergistic drug targets, Sci. Signal., .
[18]
Y. Tu, Q. Wu, J. He, J. Xu, S. Yu, Q. Wang, Y. Cheng, Q. Yang, S. Xu, and Y. Cao, Exploring the potential molecular mechanism of scutellaria baicalensis georgi in the treatment of gastric cancer based on network pharmacological analysis and molecular docking technology, Front. Pharmacol., .
[19]
L. Guo, H. Shi, and L. Zhu, Siteng fang reverses multidrug resistance in gastric cancer: A network pharmacology and molecular docking study, Front. Oncol., .
[20]
O. A. Kolawole, F. Olatomide A., and S. Banjo, Anti-gastric cancer activity of 1,2,3-triazolo[4,5-d]pyrimidine hybrids (1,2,3-TPH): QSAR and molecular docking approaches, Heliyon, .
[21]
K. Li, Y. Du, L. Li, and D. -Q. Wei, Bioinformatics approaches for anti-cancer drug discovery, Curr. Drug Targets, vol. 21, no. 1, pp. 3–17, 2019.
[22]
A. C. Kaushik, Y. J. Wang, X. Wang, and D. -Q. Wei, Irinotecan and vandetanib create synergies for treatment of pancreatic cancer patients with concomitant TP53 and KRAS mutations, Brief. Bioinform., .
[23]
M. Ö. Cingiz, G. Biricik, and B. Diri, The performance comparison of gene co-expression networks of breast and prostate cancer using different selection criteria, Interdiscip. Sci.: Comput. Life Sci., vol. 13, no. 3, pp. 500–510, 2021.
[24]
D. Szklarczyk, A. L. Gable, D. Lyon, A. Junge, S. Wyder, J. Huerta-Cepas, M. Simonovic, N. T. Doncheva, J. H. Morris, P. Bork, et al., STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets, Nucleic Acids Res., .
[25]
R. K. Patel and M. Jain, NGS QC toolkit: A toolkit for quality control of next generation sequencing data, PLoS One, .
[26]
H. Li and R. Durbin, Fast and accurate short read alignment with burrows-wheeler transform, Bioinformatics, .
[27]
Y. Liao, G. K. Smyth, and W. Shi, Featurecounts: An efficient general purpose program for assigning sequence reads to genomic features, Bioinformatics, .
[28]
Z. Li, P. Hou, D. Fan, M. Dong, M. Ma, H. Li, R. Yao, B. Huang, G. Wang, P. Geng, et al., The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer, Cell Death Differ., vol. 24, no. 1, pp. 59–71, 2017.
[29]
E. D. Tetzlaff, P. Cen, and J. A. Ajani, Emerging drugs in the treatment of advanced gastric cancer, Expert Opinion on Emerging Drugs, .
[30]
L. Vecchione, M. Orditura, F. Ciardiello, and F. D. Vita, Novel investigational drugs for gastric cancer, Expert Opinion on Investigational Drugs, .
[31]
R. Geng, L. Song, J. Li, and L. Zhao, The safety of apatinib for the treatment of gastric cancer, Expert Opin. Drug Saf., .
[32]
D. A. Hescheler, P. S. Plum, T. Zander, A. Quaas, M. Korenkov, A. Gassa, M. Michel, C. J. Bruns, and H. Alakus, Identification of targeted therapy options for gastric adenocarcinoma by comprehensive analysis of genomic data, Gastric Cancer, .
[33]
M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol., .
[34]
P. Shannon, A. Markiel, O. Ozier, N. S. Baliga, J. T. Wang, D. Ramage, N. Amin, B. Schwikowski, and T. Ideker, Cytoscape: A software environment for integrated models of biomolecular interaction networks, Genome Res., .
[35]
D. W. Huang, B. T. Sherman, and R. A. Lempicki, Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists, Nucleic Acids Res., .
[36]
Z. Tang, B. Kang, C. Li, T. Chen, and Z. Zhang, GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis, Nucleic Acids Res., .
[37]
M. V. Kuleshov, M. R. Jones, A. D. Rouillard, N. F. Fernandez, Q. Duan, Z. Wang, S. Koplev, S. L. Jenkins, K. M. Jagodnik, A. Lachmann, et al., Enrichr: A comprehensive gene set enrichment analysis web server 2016 update, Nucleic Acids Res., .
[38]
J. Pinero, J. M. Ramírez-Anguita, J. Saüch-Pitarch, F. Ronzano, E. Centeno, F. Sanz, and L. I. Furlong, The DisGeNET knowledge platform for disease genomics: 2019 update, Nucleic Acids Res., .
[39]
H. M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T. N. Bhat, H. Weissig, I. N. Shindyalov, and P. E. Bourne, The protein data bank, Nucleic Acids Research, .
[40]
A. Waterhouse, M. Bertoni, S. Bienert, G. Studer, G. Tauriello, R. Gumienny, F. T. Heer, T. A. P. D. Beer, C. Rempfer, L. Bordoli, et al., SWISS-MODEL: Homology modelling of protein structures and complexes, Nucleic Acids Res., .
[41]
S. Kim, J. Chen, T. Cheng, A. Gindulyte, J. He, S. He, Q. Li, B. A. Shoemaker, P. A. Thiessen, B. Yu, et al., PubChem 2019 update: Improved access to chemical data, Nucleic Acids Res., .
[42]
D. S. v4. 0. 100. 13345 Visualizer, Accelrys Software Inc, https://discover.3ds.com/discovery-studio-visualizer-download, 2005.
[43]
T. J. Dolinsky, P. Czodrowski, H. Li, J. E. Nielsen, J. H. Jensen, G. Klebe, and N. A. Baker, PDB2PQR: Expanding and upgrading automated preparation of biomolecular structures for molecular simulations, Nucleic Acids Res., .
[44]
J. C. Gordon, J. B. Myers, T. Folta, V. Shoja, L. S. Heath, and A. Onufriev, H++: A server for estimating pKas and adding missing hydrogens to macromolecules, Nucleic Acids Res., .
[45]
G. M. Morris, R. Huey, W. Lindstrom, M. F. Sanner, R. K. Belew, D. S. Goodsell, and A. J. Olson, AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility, J. Comput. Chem., vol. 30, no. 16, pp. 2785–2791, 2009.
[46]
O. Trott and A. J. Olson, AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading, J. Comput. Chem., vol. 31, no. 2, pp. 455–461, 2010.
[47]
W. L. Delano and S. Bromberg, PyMOL user’s guide, https://pymol.sourceforge.net/newman/userman.pdf, 2004.
[48]
W. L. DeLano, The PyMOL molecular graphics system, version 2.3, Schrödinger LLC, https://sourceforge.net/projects/pymol/, 2020.
[49]
D506-D515, UniProt: A worldwide hub of protein knowledge, The UniProt Consortium, Nucleic Acids Res., https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6323992/, 2019.
[50]
O. M. Omar, M. Soutto, N. S. Bhat, A. A. Bhat, H. Lu, Z. Chen, and W. El-Rifai, TFF1 antagonizes TIMP-1 mediated proliferative functions in gastric cancer, Mol. Carcinog., vol. 57, no. 11, pp. 1577–1587, 2018.
[51]
W. Chen and Z. Yang, Identification of differentially expressed genes reveals BGN predicting overall survival and tumor immune infiltration of gastric cancer, Comput. Math. Methods Med., .
[52]
T. Chen, L. Sun, C. He, Y. Gong, Q. Xu, and Y. Yuan, Serum OPN expression for identification of gastric cancer and atrophic gastritis and its influencing factors, PLoS One, .
[53]
D. Oikonomou, K. Hassan, J. T. Kaifi, H. C. Fiegel, P. G. Schurr, U. Reichelt, K. Aridome, E. F. Yekebas, O. Mann, D. Kluth, et al., Thy-1 as a potential novel diagnostic marker for gastrointestinal stromal tumors, J. Cancer Res. Clin. Oncol., vol. 133, no. 12, pp. 951–955, 2007.
[54]
Y. C. Chuang, H. -Y. Wu, Y. -L. Lin, S. -C. Tzou, C. -H. Chuang, T. -Y. Jian, P. -R. Chen, Y. -C. Chang, C. -H. Lin, T. -H. Huang, et al., Blockade of ITGA2 induces apoptosis and inhibits cell migration in gastric cancer, Biol. Proced. Online, .
[55]
S. Soleyman-Jahi, S. Nedjat, A. Abdirad, N. Hoorshad, R. Heidari, and K. Zendehdel, Prognostic significance of matrix metalloproteinase-7 in gastric cancer survival: A meta-analysis, PLoS One, .
[56]
J. Qiu, M. Sun, Y. Wang, and B. Chen, Identification of hub genes and pathways in gastric adenocarcinoma based on bioinformatics analysis, Med. Sci. Monit., .
[57]
B. Jiang, S. Li, Z. Jiang, and P. Shao, Gastric cancer associated genes identified by an integrative analysis of gene expression data, Biomed Res. Int., .
[58]
X. Chong, R. Peng, Y. Sun, L. Zhang, and Z. Zhang, Identification of key genes in gastric cancer by bioinformatics analysis, Biomed Res. Int., .
[59]
W. Wang, Y. He, Q. Zhao, X. Zhao, and Z. Li, Identification of potential key genes in gastric cancer using bioinformatics analysis, Biomed. Reports, vol. 12, no. 4, pp. 178–192, 2020.
[60]
K. Hu and F. Chen, Identification of significant pathways in gastric cancer based on protein-protein interaction networks and cluster analysis, Genet. Mol. Biol., vol. 35, no. 3, pp. 701–708, 2012.
[61]
D. Mao, R. Xu, H. Chen, X. Chen, D. Li, S. Song, Y. He, Z. Wei, and C. Zhang, Cross-talk of focal adhesion-related gene defines prognosis and the immune microenvironment in gastric cancer, Front. Cell Dev. Biol., .
[62]
T. Matsuoka and M. Yashiro, The role of PI3K/Akt/mTOR signaling in gastric carcinoma, Cancers, vol. 6, no. 3, pp. 1441–1463, 2014.
[63]
S. Luo, R. Lin, X. Liao, D. Li, and Y. Qin, Identification and verification of the molecular mechanisms and prognostic values of the cadherin gene family in gastric cancer, Sci. Rep., vol. 11, no. 1, p. 23674, 2021.
[64]
R. Huang, W. Gu, B. Sun, and L. Gao, Identification of COL4A1 as a potential gene conferring trastuzumab resistance in gastric cancer based on bioinformatics analysis, Mol. Med. Rep., vol. 17, no. 5, pp. 6387–6396, 2018.
[65]
Y. L. Ding, S. F. Sun, and G. L. Zhao, COL5A2 as a potential clinical biomarker for gastric cancer and renal metastasis, Medicine (Baltimore)., vol. 100, no. 7, p. e24561, 2021.
[66]
X. Liu and S. J. Meltzer, Gastric cancer in the era of precision medicine, CMGH, vol. 3, no. 3. pp. 348–358, 2017.
[67]
G. A. Colditz, National cancer institute, SAGE Encycl. Cancer Soc., .
[68]
T. Lim, J. Lee, D. J. Lee, H. Y. Lee, B. Han, K. K. Baek, H. K. Ahn, S. J. Lee, S. H. Park, J. O. Park, et al., Phase I trial of capecitabine plus everolimus (RAD001) in patients with previously treated metastatic gastric cancer, Cancer Chemother. Pharmacol., vol. 68, no. 1, pp. 255–262, 2011.
[69]
C. Pauligk, S. Janowski, K. Steinmetz, A. Atmaca, A. Pustowka, E. Jaeger, and S. Al-Batran, Daily RAD001 plus mitomycin C, every three weeks in previously treated patients with advanced gastric cancer or cancer of the esophagogastric junction: Preliminary results of a phase I study, J. Clin. Oncol., vol. 28, no. 15_suppl, p. e14531, 2010.
[70]
T. Doi, K. Muro, N. Boku, Y. Yamada, T. Nishina, H. Takiuchi, Y. Komatsu, Y. Hamamoto, N. Ohno, and Y. Fujita, Multicenter phase II study of everolimus in patients with previously treated metastatic gastric cancer, J. Clin. Oncol., vol. 28, no. 11, pp. 1904–1910, 2010.
[71]
E. V. Cutsem, C. Boni, J. Tabernero, B. Massuti, G. Middleton, F. Dane, P. Reichardt, F. L. Pimentel, A. Cohn, P. Follana, et al., Docetaxel plus oxaliplatin with or without fluorouracil or capecitabine in metastatic or locally recurrent gastric cancer: A randomized phase II study, Ann. Oncol., vol. 26, no. 1, pp. 149–156, 2015.
[72]
H. Lin, D. Han, G. Fu, C. Liu, L. Wang, S. Han, B. Liu, and J. Yu, Concurrent apatinib and docetaxel vs apatinib monotherapy as third-or subsequent-line therapy for advanced gastric adenocarcinoma: A retrospective study, Onco. Targets. Ther., vol. 12, pp. 1681–1689, 2019.
[73]
E. M. Wolin, A. Manon, C. Chassaing, A. Lewis, L. Bertocchi, J. Richard, and A. T. Phan, Lanreotide depot: An antineoplastic treatment of carcinoid or neuroendocrine tumors, Journal of Gastrointestinal Cancer, vol. 47, no. 4. pp. 366–374, 2016.
[74]
A. J. Souers, J. D. Leverson, E. R. Boghaert, S. L. Ackler, N. D. Catron, J. Chen, B. D. Dayton, H. Ding, S. H. Enschede, W. J. Fairbrother, et al., ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets, Nat. Med., vol. 19, no. 2, pp. 202–208, 2013.
[75]
H. Fukamachi, S. -K. Kim, J. Koh, H. S. Lee, Y. Sasaki, K. Yamashita, T. Nishikawaji, S. Shimada, Y. Akiyama, S. J. Byeon, et al., A subset of diffuse-type gastric cancer is susceptible to mTOR inhibitors and checkpoint inhibitors, J. Exp. Clin. Cancer Res., .
[76]
S. J. Byeon, N. Han, J. Choi, M. A. Kim, and W. H. Kim, Prognostic implication of TSC1 and mTOR expression in gastric carcinoma, J. Surg. Oncol., vol. 109, no. 8, pp. 812–817, 2014.
[77]
S. E. Al-Batran, M. Ducreux, and A. Ohtsu, MTOR as a therapeutic target in patients with gastric cancer, International Journal of Cancer, vol. 130, no. 3. pp. 491–496, 2012.
[78]
T. Murayama, M. Inokuchi, Y. Takagi, H. Yamada, K. Kojima, J. Kumagai, T. Kawano, and K. Sugihara, Relation between outcomes and localisation of p-mTOR expression in gastric cancer, Br. J. Cancer, vol. 100, no. 5, pp. 782–788, 2009.
[79]
T. Huang, F. Zhou, X. Yuan, T. Yang, X. Liang, Y. Wang, H. Tu, J. Chang, K. Nan, and Y. Wei, Reactive oxygen species are involved in the development of gastric cancer and gastric cancer-related depression through ABL1-mediated inflammation signaling pathway, Oxid. Med. Cell. Longev., .
[80]
N. Kassem, O. M. Ismail, H. Elomri, and M. A. Yassin, Nilotinib induced recurrent gastric polyps: Case report and review of literature, American Journal of Case Reports, vol. 18, pp. 794–798, 2017.
File
649-664ESM.pdf (130.4 KB)
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 18 February 2022
Revised: 31 May 2022
Accepted: 29 August 2022
Published: 06 January 2023
Issue date: August 2023

Copyright

© The author(s) 2023.

Acknowledgements

This work was partly supported by the National Key Research and Development Program of China (No. 2018YFB0204403), Key Research and Development Project of Guangdong Province (No. 2021B0101310002), Strategic Priority CAS Project (No. XDB38050100), National Science Foundation of China (No. U1813203), the Shenzhen Basic Research Fund (Nos. RCYX2020071411473419, KQTD20200820113106007, and JSGG20201102163800001), CAS Key Lab (No. 2011DP173015), and the Youth Innovation Promotion Association (No. Y2021101).

We would also like to thank all the members of Computational Biology and Bioinformatics Lab, Center for High Performance Computing, SIAT, CAS for their valuable suggestions and feed-backs.

Rights and permissions

The articles published in this open access journal are distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/).

Return