Amoeboid cells undergo durotaxis with soft end polarized NMIIA

  1. Institute of Systems Biomedicine, Schol of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
  2. Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191 China;
  3. International Cancer Institute, Peking University, Beijing, 100191 China;
  4. Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
  5. National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China
  6. School of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, China
  7. College of Life Sciences, University of Chinese Academy of Sciences, 100049 Beijing, China

Peer review process

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

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Yi Arial Zeng
    Chinese Academy of Sciences, Shanghai, China
  • Senior Editor
    Jonathan Cooper
    Fred Hutchinson Cancer Research Center, Seattle, United States of America

Reviewer #1 (Public Review):

In their paper, Kang et al. investigate rigidity sensing in amoeboid cells, showing that, despite their lack of proper focal adhesions, amoeboid migration of single cells is impacted by substrate rigidity. In fact, many different amoeboid cell types can durotax, meaning that they preferentially move towards the stiffer side of a rigidity gradient.

The authors observed that NMIIA is required for durotaxis and, building on this observation, they generated a model to explain how durotaxis could be achieved in the absence of strong adhesions. According to the model, substrate stiffness alters the diffusion rate of NMAII, with softer substrates allowing for faster diffusion. This allows for NMAII accumulation at the back, which, in turn, results in durotaxis.

The experiments support the main message of the paper regarding durotaxis by amoeboid cells. In my opinion, a few clarifications on the mechanism proposed to explain this phenomenon could strengthen this research:

(1) According to your model, the rear end of the cell, which is in contact with softer substrates, will have slower diffusion rates of MNIIA. Does this mean that bigger cells will durotax better than smaller cells because the stiffness difference between front and rear is higher? Is it conceivable to attenuate the slope of the durotactic gradient to a degree where smaller cells lose their ability to durotact, while longer cells retain their capacity for directional movement?

(2) Where did you place the threshold for soft, middle, and stiff regions (Figure 6)? Is it possible that you only have a linear rigidity gradient in the center of your gel and the more you approach the borders, the flatter the gradient gets? In this case, cells would migrate randomly on uniform substrates. Did you perform AFM over the whole length of the gel or just in the central part?

(3) In which region (soft, middle, stiff) did you perform all the cell tracking of the previous figures?

(4) What is the level of confinement experienced by the cells? Is it possible that cells on the soft side of the gels experience less confinement due to a "spring effect" whereby the coverslips descending onto the cells might exert diminished pressure because the soft hydrogels act as buffers, akin to springs? If this were the case, cells could migrate following a confinement gradient.

Reviewer #2 (Public Review):

Summary:
The authors developed an imaging-based device that provides both spatial confinement and stiffness gradient to investigate if and how amoeboid cells, including T cells, neutrophils, and Dictyostelium, can durotax. Furthermore, the authors showed that the mechanism for the directional migration of T cells and neutrophils depends on non-muscle myosin IIA (NMIIA) polarized towards the soft-matrix-side. Finally, they developed a mathematical model of an active gel that captures the behavior of the cells described in vitro.

Strengths:

The topic is intriguing as durotaxis is essentially thought to be a direct consequence of mechanosensing at focal adhesions. To the best of my knowledge, this is the first report on amoeboid cells that do not depend on FAs to exert durotaxis. The authors developed an imaging-based durotaxis device that provides both spatial confinement and stiffness gradient and they also utilized several techniques such as quantitative fluorescent speckle microscopy and expansion microscopy. The results of this study have well-designed control experiments and are therefore convincing.

Weaknesses:

Overall this study is well performed but there are still some minor issues I recommend the authors address:

(1) When using NMIIA/NMIIB knockdown cell lines to distinguish the role of NMIIA and NMIIB in amoeboid durotaxis, it would be better if the authors took compensatory effects into account.
(2) The expansion microscopy assay is not clearly described and some details are missed such as how the assay is performed on cells under confinement.
(3) In this study, an active gel model was employed to capture experimental observations. Previously, some active nematic models were also considered to describe cell migration, which is controlled by filament contraction. I suggest the authors provide a short discussion on the comparison between the present theory and those prior models.
(4) In the present model, actin flow contributes to cell migration while myosin distribution determines cell polarity. How does this model couple actin and myosin together?

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