De novo identification of universal cell mechanics gene signatures

  1. Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, Dresden 01307, Germany
  2. Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Staudtstraße 2, Erlangen 91058, Germany
  3. Department of Computer Science and Department of Economics, Jamia Millia Islamia, New Delhi 110025, India
  4. Center for Complex Network Intelligence, Tsinghua Laboratory of Brain and Intelligence, Department of Computer Science and Department of Biomedical Engineering, Tsinghua University, Chengfu Rd. 160, SanCaiTang Building, Haidian District, Beijing 100084, China
  5. Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Fetscherstrasse 105, Dresden 01307, Germany
  6. Klinik und Poliklinik für Kinder-und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, Dresden 01307, Germany
  7. Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
  8. Mechanoadaptation and Caveolae Biology lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle de Melchor Fernández Almagro 3, Madrid 28029, Spain
  9. Center for Systems Biology Dresden, Pfotenhauerstraße 108, Dresden 01307, Germany
  10. Cluster of Excellence Physics of Life, Technische Universität Dresden, Tatzberg 47-49, Dresden 01307, Germany

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Ahmad Khalil
    Boston University, Boston, United States of America
  • Senior Editor
    Aleksandra Walczak
    École Normale Supérieure - PSL, Paris, France

Reviewer #1 (Public Review):

In this work, Urbanska and colleagues use a machine-learning based crossing of mechanical characterisations of various cells in different states and their transcriptional profiles. Using this approach, they identify a core set of five genes that systematically vary together with the mechanical state of the cells, although not always in the same direction depending on the conditions. They show that the combined transcriptional changes in this gene set is strongly predictive of a change in the cell mechanical properties, in systems that were not used to identify the genes (a validation set). Finally, they experimentally after the expression level of one of these genes, CAV1, that codes for the caveolin 1 protein, and show that, in a variety of cellular systems and contexts, perturbations in the expression level of CAV1 also induce changes in cell mechanics, cells with lower CAV1 expression being generally softer.

Overall the approach seems accessible, sound and is well described. My personal expertise is not suited to judge its validity, novelty or relevance, so I do not make comments on that. The results it provides seem to have been thoroughly tested by the authors (using different types of mechanical characterisations of the cells) and to be robust in their predictive value. The authors also show convincingly that one of the genes they identified, CAV1, is not only correlated with the mechanical properties of cells, but also that changing its expression level affects cell mechanics. At this stage, the study appears mostly focused on the description and validation of the methodological approach, and it is hard to really understand what the results obtain really mean, the importance of the biological finding - what is this set of 5 genes doing in the context of cell mechanics? Is it really central, or is it just one of the set of knobs on which the cell plays - and it is identified by this method because it is systematically modulated but maybe, for any given context, it is not the dominant player - all these fundamental questions remain unanswered at this stage. On one hand, it means that the study might have identified an important novel module of genes in cell mechanics, but on the other hand, it also reveals that it is not yet easy to interpret the results provided by this type of novel approach.

Reviewer #2 (Public Review):

A key strength is the quantitative approaches all add rigor to what is being attempted. The approach with very different cell culture lines will in principle help identify constitutive genes that vary in a particular and predictable way. To my knowledge, one other study that should be cited posed a similar pan-tissue question using mass spectrometry proteomics instead of gene expression, and also identified a caveolae component (cavin-1, PTRF) that exhibited a trend with stiffness across all sampled tissues. The study focused instead on a nuclear lamina protein that was also perturbed in vitro and shown to follow the expected mechanical trend (Swift et al 2013).

Reviewer #3 (Public Review):

In this work, Urbanska et al. link the mechanical phenotypes of human glioblastoma cell lines and murine iPSCs to their transcriptome, and using machine learning-based network analysis identify genes with putative roles in cell mechanics regulation. The authors identify 5 target genes whose transcription creates a combinatorial marker which can predict cell stiffness in human carcinoma and breast epithelium cell lines as well as in developing mouse neurons. For one of the target genes, caveolin1 (CAV1), the authors perform knockout, knockdown, overexpression and rescue experiments in human carcinoma and breast epithelium cell lines. They determine the cell stiffness via RT-DC, AFM indentation and AFM rheology and confirm that high CAV1 expression levels correlate with increased stiffness in those model systems. This work brings forward an interesting approach to identify novel genes in an unbiased manner, but surprisingly the authors validate caveolin 1, a target gene with known roles in cell mechanics regulation.

I have two main concerns with the current version of this work:

  1. The authors identify a network of 5 genes that can predict mechanics. What is the relationship between the 5 genes? If the authors aim to highlight the power of their approach by knockdown, knockout or over-expression of a single gene why choose CAV1 (which has an individual p-value of 0.16 in Fig S4)? To justify their choice, the authors claim that there is limited data supporting the direct impact of CAV1 on mechanical properties of cells but several studies have previously shown its role in for example zebrafish heart stiffness, where a knockout leads to higher stiffness (Grivas et al., Scientific Reports 2020), in cancer cells, where a knockdown leads to cell softening (Lin et al., Oncotarget 2015), or in endothelial cell, where a knockout leads to cell softening (Le Master et al., Scientific Reports 2022).
  2. The authors do not show how much does PC-Corr outperforms classical co-expression network analysis or an alternative gold standard. It is worth noting that PC-Corr was previously published by the same authors to infer phenotype-associated functional network modules from omics datasets (Ciucci et al., Scientific Reports 2017).

Altogether, the authors provide an interesting approach to identify novel genes associated with cell mechanics changes, but the current version does not fulfill such potential by focusing on a single gene with known roles in cell mechanics.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation