Next Article in Journal
Genome Assessment of Carbapenem- and Colistin-Resistant Escherichia coli from Patients in a Sentinel Hospital in China
Previous Article in Journal
Fabricated AIE-Based Probe to Detect the Resistance to Anoikis of Cancer Cells Detached from Tumor Tissue
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Cutting-Edge Methods for Better Understanding Cells

Department of Bioinformatics and Systems Biology, MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
Cells 2022, 11(21), 3479; https://doi.org/10.3390/cells11213479
Submission received: 31 October 2022 / Accepted: 1 November 2022 / Published: 3 November 2022
(This article belongs to the Section Cell Methods)
Cells are microscopic yet fundamental elements of life. The in-depth analysis of the regulatory mechanisms and phenotypic dynamics in cells is the foundation of understanding human health and diseases. Thus, the development of novel, efficient, accurate and low-cost methods has been highly encouraged for analyzing cells. In this book, I am happy to present 12 prominent papers published in Cells from 2020 to 2021. The methods reported in these papers covered cutting-edge techniques in single-cell analysis, gene expression data analysis, biological sequence analysis, drug discovery and genetic and/or imaging analyses.
For the single-cell analysis, five papers were selected, covering single-cell morphological monitoring, the isolation of single T cells, deep-learning-based analysis of single-cell RNA-seq (scRNA-seq) data, cross-species analysis of scRNA-seq data and validation of scRNA-seq-derived results. For the ultra-high-throughput monitoring of the morphology of individual red blood cells (RBCs), Park et al. developed a holographic cytometry instrument. Coupled with machine learning algorithms, such as logistic regression, they demonstrated that morphological changes to RBCs could be accurately measured and quantified [1]. Weiss et al. carefully tested three commonly used methods for the isolation of CD3-specific T cells from human buffy coats and peripheral blood mononuclear cells (PBMCs) based on the rationale of either immune affinity chromatography or magnetic beads. They found all three methods to be highly efficient for isolating T cells, with a high viability (>92%) and purity (>98%) [2]. For the data analysis, Walbech et al. implemented a deep learning framework of autoencoders to extract informative features from scRNA-seq data, as well as for further denoising and classification. They suggested that such a method could be useful for the identification of novel cell types not included in the training dataset [3]. Additionally, Gao et al. profiled a comprehensive transcriptomic atlas of human and mouse hematopoietic stem and progenitor cells (HSPCs), with a total of 32,805 single cells. They used a number of scRNA-seq analysis tools, such as Monocle, Seurat 2 and SCENIC, to analyze the data. Through conducting a comparison, they identified conserved HSPC types, conserved cell-type expression profiles and conserved cell-type-specific regulatory elements, motifs and networks [4]. For the validation of scRNA-seq results, Zucha et al. provided a step-by-step tutorial of a reverse transcription quantitative PCR (RT-qPCR), which could also be used for efficient screening prior to scRNA-Seq experiments. This tutorial covered sample collection, reverse transcription, preamplification, quantitative PCR and the data analysis [5].
For the gene expression data analysis, Tao et al. reanalyzed 2296 gene microarray datasets in Caenorhabditis elegans, and identified rps-23, the 40S ribosomal protein S23, as a critical housekeeping gene that could be used as a potential reference for the reliable normalization and quantification of gene expression data [6]. Esmaeili et al. performed a meta-analysis of six published bulk RNA sequencing (RNA-seq) data, and implemented three types of machine learning algorithms to identify potentially key transcription factors (TFs) to be regulated using herbal compounds in cancer cells. Using this approach, they identified four TFs, including AIP, VGLL4, TFE3 and ID1, to be differentially expressed with the treatment of genistein, an isoflavone compound found in soy products and in liver cancer HepG2 cells [7].
For the biological sequence analysis, Wahab et al. encoded DNA sequences with each of the six types of features, such as the dinucleotide composition (DNC) and trinucleotide composition (TNC). Then, they used a convolution neural network (CNN) to develop a predictor, DNC4mC-Deep, for the prediction of potential N4-methylcytosine (4mC) in two plant genomes, Fragaria vesca and Rosa vhinensis, with the area under the ROC curve (AUC) value ranging from 0.90 to 0.96 [8].
Two papers were selected for drug discovery. First, Benchoua et al. provided a comprehensive review of various types of brain cells derived from human pluripotent stem cells (PSCs), and carefully demonstrated how these cell models were greatly helpful for the discovery of therapeutic drugs for rare genetic disorders, neurodegenerative diseases and psychiatric disorders [9]. Additionally, Dang et al. encoded small molecules using simplified molecular-input line-entry systems (SMILESs) and chemical interaction features on the CYP450 group, and developed a machine-learning-based method, histamine antagonist interaction-network inference (HAINI), by testing a variety of classification algorithms, such as the decision tree, with an AUC value of 0.96 [10].
Moreover, I selected two papers concerning genetic and imaging analyses. Kondratyev et al. reviewed the cutting-edge progress achieved in genome-wide association studies (GWASs) for the identification of genetic factors to interpret complex traits. They introduced the basic concepts and strategies in GWASs, elaborated their pros and cons, and emphasized the importance of the appropriate interpretation of GWAS findings for experimental biologists [11]. Wu et al. reviewed recent progress in using machine learning methods for the analysis of imaging and genomic data of gliomas. Three important aspects, including biomarker prediction, grade classification and prognostic prediction, were summarized. Challenges in this field, such as the lack of annotated data, data class imbalances and model overfitting, were also discussed [12].
As the Editor-in-Chief of the “Cell Methods” Section of the Cells journal, I am glad to have chosen these representative studies, providing leading methods or methodological reviews for Cells readers. For the future improvement of this section, I hope to share my thoughts here. First, the Cell Methods Section highly encourages all submissions covering the development of bioimaging, computational, bioinformatics and/or artificial intelligence methods with the capacity to help study cells. For convenience, at least one application in cells should be performed to demonstrate how the novel method can advance our understanding of cells. If possible, a brief summary on the usefulness of the method in cells should be included in the cover letter, prior to submission. I believe such a procedure would facilitate the decision making by the editorial office, as well as the following external review. Additionally, biologists working on cells would also find it easier to learn the importance of the reported methods.

Funding

This study was funded by National Key R & D Program of China (2021YFF0702000, 2022YFC2704300, and 2021ZD0201300), Natural Science Foundation of China (31930021 and 31970633), and Hubei Innovation Group Project (2021CFA005).

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Park, H.S.; Price, H.; Ceballos, S.; Chi, J.T.; Wax, A. Single cell analysis of stored red blood cells using ultra-high throughput holographic cytometry. Cells 2021, 10, 2455. [Google Scholar] [CrossRef] [PubMed]
  2. Weiss, R.; Gerdes, W.; Berthold, R.; Sack, U.; Koehl, U.; Hauschildt, S.; Grahnert, A. Comparison of three cd3-specific separation methods leading to labeled and label-free t cells. Cells 2021, 10, 2824. [Google Scholar] [CrossRef] [PubMed]
  3. Walbech, J.S.; Kinalis, S.; Winther, O.; Nielsen, F.C.; Bagger, F.O. Interpretable autoencoders trained on single cell sequencing data can transfer directly to data from unseen tissues. Cells 2021, 11, 85. [Google Scholar] [CrossRef] [PubMed]
  4. Gao, S.; Wu, Z.; Kannan, J.; Mathews, L.; Feng, X.; Kajigaya, S.; Young, N.S. Comparative transcriptomic analysis of the hematopoietic system between human and mouse by single cell rna sequencing. Cells 2021, 10, 973. [Google Scholar] [CrossRef] [PubMed]
  5. Zucha, D.; Kubista, M.; Valihrach, L. Tutorial: Guidelines for single-cell rt-qpcr. Cells 2021, 10, 2607. [Google Scholar] [CrossRef] [PubMed]
  6. Tao, J.; Hao, Y.; Li, X.; Yin, H.; Nie, X.; Zhang, J.; Xu, B.; Chen, Q.; Li, B. Systematic identification of housekeeping genes possibly used as references in caenorhabditis elegans by large-scale data integration. Cells 2020, 9, 786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Esmaeili, F.; Lohrasebi, T.; Mohammadi-Dehcheshmeh, M.; Ebrahimie, E. Evaluation of the effectiveness of herbal components based on their regulatory signature on carcinogenic cancer cells. Cells 2021, 10, 3139. [Google Scholar] [CrossRef] [PubMed]
  8. Wahab, A.; Mahmoudi, O.; Kim, J.; Chong, K.T. Dnc4mc-deep: Identification and analysis of DNA n4-methylcytosine sites based on different encoding schemes by using deep learning. Cells 2020, 9, 1756. [Google Scholar] [CrossRef] [PubMed]
  9. Benchoua, A.; Lasbareilles, M.; Tournois, J. Contribution of human pluripotent stem cell-based models to drug discovery for neurological disorders. Cells 2021, 10, 3290. [Google Scholar] [CrossRef] [PubMed]
  10. Dang, L.H.; Dung, N.T.; Quang, L.X.; Hung, L.Q.; Le, N.H.; Le, N.T.N.; Diem, N.T.; Nga, N.T.T.; Hung, S.H.; Le, N.Q.K. Machine learning-based prediction of drug-drug interactions for histamine antagonist using hybrid chemical features. Cells 2021, 10, 3092. [Google Scholar] [CrossRef] [PubMed]
  11. Kondratyev, N.V.; Alfimova, M.V.; Golov, A.K.; Golimbet, V.E. Bench research informed by gwas results. Cells 2021, 10, 3184. [Google Scholar] [CrossRef] [PubMed]
  12. Wu, Y.; Guo, Y.; Ma, J.; Sa, Y.; Li, Q.; Zhang, N. Research progress of gliomas in machine learning. Cells 2021, 10, 3169. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xue, Y. Cutting-Edge Methods for Better Understanding Cells. Cells 2022, 11, 3479. https://doi.org/10.3390/cells11213479

AMA Style

Xue Y. Cutting-Edge Methods for Better Understanding Cells. Cells. 2022; 11(21):3479. https://doi.org/10.3390/cells11213479

Chicago/Turabian Style

Xue, Yu. 2022. "Cutting-Edge Methods for Better Understanding Cells" Cells 11, no. 21: 3479. https://doi.org/10.3390/cells11213479

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