1
|
Lessenger AT, Skotheim JM, Swaffer MP, Feldman JL. Somatic polyploidy supports biosynthesis and tissue function by increasing transcriptional output. J Cell Biol 2025; 224:e202403154. [PMID: 39652010 PMCID: PMC11627111 DOI: 10.1083/jcb.202403154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 09/27/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Cell size and biosynthetic capacity generally increase with increased DNA content. Somatic polyploidy has therefore been proposed to be an adaptive strategy to increase cell size in specialized tissues with high biosynthetic demands. However, if and how DNA concentration limits cellular biosynthesis in vivo is not well understood. Here, we show that polyploidy in the Caenorhabditis elegans intestine is critical for cell growth and yolk biosynthesis, a central role of this organ. Artificially lowering the DNA/cytoplasm ratio by reducing polyploidization in the intestine gave rise to smaller cells with dilute mRNA. Highly expressed transcripts were more sensitive to this mRNA dilution, whereas lowly expressed genes were partially compensated-in part by loading more RNA Polymerase II on the remaining genomes. Polyploidy-deficient animals produced fewer and slower-growing offspring, consistent with reduced synthesis of highly expressed yolk proteins. DNA-dilute cells had normal total protein concentration, which we propose is achieved by increasing the expression of translational machinery at the expense of specialized, cell-type-specific proteins.
Collapse
Affiliation(s)
| | - Jan M. Skotheim
- Department of Biology, Stanford University, Stanford, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Mathew P. Swaffer
- Department of Biology, Stanford University, Stanford, CA, USA
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
2
|
Liu S, Tan C, Melo-Gavin C, Ginzberg MB, Blutrich R, Patel N, Rape M, Mark KG, Kafri R. Oversized cells activate global proteasome-mediated protein degradation to maintain cell size homeostasis. eLife 2025; 14:e75393. [PMID: 39791360 PMCID: PMC11810107 DOI: 10.7554/elife.75393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
Proliferating animal cells maintain a stable size distribution over generations despite fluctuations in cell growth and division size. Previously, we showed that cell size control involves both cell size checkpoints, which delay cell cycle progression in small cells, and size-dependent regulation of mass accumulation rates (Ginzberg et al., 2018). While we previously identified the p38 MAPK pathway as a key regulator of the mammalian cell size checkpoint (Liu et al., 2018), the mechanism of size-dependent growth rate regulation has remained elusive. Here, we quantified global rates of protein synthesis and degradation in cells of varying sizes, both under unperturbed conditions and in response to perturbations that trigger size-dependent compensatory growth slowdown. We found that protein synthesis rates scale proportionally with cell size across cell cycle stages and experimental conditions. In contrast, oversized cells that undergo compensatory growth slowdown exhibit a superlinear increase in proteasome-mediated protein degradation, with accelerated protein turnover per unit mass, suggesting activation of the proteasomal degradation pathway. Both nascent and long-lived proteins contribute to the elevated protein degradation during compensatory growth slowdown, with long-lived proteins playing a crucial role at the G1/S transition. Notably, large G1/S cells exhibit particularly high efficiency in protein degradation, surpassing that of similarly sized or larger cells in S and G2, coinciding with the timing of the most stringent size control in animal cells. These results collectively suggest that oversized cells reduce their growth efficiency by activating global proteasome-mediated protein degradation to promote cell size homeostasis.
Collapse
Affiliation(s)
- Shixuan Liu
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Cell Biology, The Hospital for Sick Children, TorontoTorontoCanada
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Ceryl Tan
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Cell Biology, The Hospital for Sick Children, TorontoTorontoCanada
| | - Chloe Melo-Gavin
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Cell Biology, The Hospital for Sick Children, TorontoTorontoCanada
| | | | - Ron Blutrich
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Cell Biology, The Hospital for Sick Children, TorontoTorontoCanada
| | - Nish Patel
- Cell Biology, The Hospital for Sick Children, TorontoTorontoCanada
| | - Michael Rape
- Department of Molecular Cell Biology, University of California at BerkeleyBerkeleyUnited States
| | - Kevin G Mark
- Department of Molecular Cell Biology, University of California at BerkeleyBerkeleyUnited States
- Department of Cell Biology, UT Southwestern Medical CenterDallasUnited States
| | - Ran Kafri
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Cell Biology, The Hospital for Sick Children, TorontoTorontoCanada
| |
Collapse
|
3
|
Fu J, Ni Q, Wu Y, Gupta A, Ge Z, Yang H, Afrida Y, Barman I, Sun S. Cells Prioritize the Regulation of Cell Mass Density. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627803. [PMID: 39713365 PMCID: PMC11661194 DOI: 10.1101/2024.12.10.627803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
A cell's global physical state is characterized by its volume and dry mass. The ratio of cell mass to volume is the cell mass density (CMD), which is also a measure of macromolecular crowding and concentrations of all proteins. Using the Fluorescence eXclusion method (FXm) and Quantitative Phase Microscopy (QPM), we investigate CMD dynamics after exposure to sudden media osmolarity change. We find that while the cell volume and mass exhibit complex behavior after osmotic shock, CMD follows a straightforward monotonic recovery in 48 hours. The recovery is cell-cycle independent and relies on a coordinated adjustment of protein synthesis and volume growth rates. Surprisingly, we find that the protein synthesis rate decreases when CMD increases. This result is explained by CMD-dependent nucleoplasm-cytoplasm transport, which serves as negative regulatory feedback on CMD. The Na+/H+ exchanger NHE plays a role in regulating CMD by affecting both protein synthesis and volume change. Taken together, we reveal that cells possess a robust control system that actively regulates CMD during environmental change.
Collapse
|
4
|
Lanz MC, Zhang S, Swaffer MP, Ziv I, Götz LH, Kim J, McCarthy F, Jarosz DF, Elias JE, Skotheim JM. Genome dilution by cell growth drives starvation-like proteome remodeling in mammalian and yeast cells. Nat Struct Mol Biol 2024; 31:1859-1871. [PMID: 39048803 DOI: 10.1038/s41594-024-01353-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
Cell size is tightly controlled in healthy tissues and single-celled organisms, but it remains unclear how cell size influences physiology. Increasing cell size was recently shown to remodel the proteomes of cultured human cells, demonstrating that large and small cells of the same type can be compositionally different. In the present study, we utilize the natural heterogeneity of hepatocyte ploidy and yeast genetics to establish that the ploidy-to-cell size ratio is a highly conserved determinant of proteome composition. In both mammalian and yeast cells, genome dilution by cell growth elicits a starvation-like phenotype, suggesting that growth in large cells is restricted by genome concentration in a manner that mimics a limiting nutrient. Moreover, genome dilution explains some proteomic changes ascribed to yeast aging. Overall, our data indicate that genome concentration drives changes in cell composition independently of external environmental cues.
Collapse
Affiliation(s)
- Michael C Lanz
- Department of Biology, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA.
| | - Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Inbal Ziv
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | - Jacob Kim
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Frank McCarthy
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Joshua E Elias
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA
| | - Jan M Skotheim
- Department of Biology, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Tan C, Lanz MC, Swaffer M, Skotheim J, Chang F. Intracellular diffusion in the cytoplasm increases with cell size in fission yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.613766. [PMID: 39386641 PMCID: PMC11463555 DOI: 10.1101/2024.09.21.613766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Diffusion in the cytoplasm can greatly impact cellular processes, yet regulation of macromolecular diffusion remains poorly understood. There is increasing evidence that cell size affects the density and macromolecular composition of the cytoplasm. Here, we studied whether cell size affects diffusion at the scale of macromolecules tens of microns in diameter. We analyzed the diffusive motions of intracellular genetically-encoded multimeric 40 nm nanoparticles (cytGEMs) in the cytoplasm of the fission yeast Schizosaccharomyces pombe . Using cell size mutants, we showed that cytGEMs diffusion coefficients decreased in smaller cells and increased in larger cells. This increase in diffusion in large cells may be due to a decrease in the DNA-to-Cytoplasm ratio, as diffusion was not affected in large multinucleate cytokinesis mutants. In investigating the underlying causes of altered cytGEMs diffusion, we found that the proteomes of large and small cells exhibited size-specific changes, including the sub-scaling of ribosomal proteins in large cells. Comparison with a similar dataset from human cells revealed that features of size-dependent proteome remodeling were conserved. These studies demonstrate that cell size is an important parameter in determining the biophysical properties and the composition of the cytoplasm.
Collapse
|
6
|
Glenn S, Fragasso A, Lin WH, Papagiannakis A, Kato S, Jacobs-Wagner C. Coupling of cell growth modulation to asymmetric division and cell cycle regulation in Caulobacter crescentus. Proc Natl Acad Sci U S A 2024; 121:e2406397121. [PMID: 39361646 PMCID: PMC11474046 DOI: 10.1073/pnas.2406397121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
In proliferating bacteria, growth rate is often assumed to be similar between daughter cells. However, most of our knowledge of cell growth derives from studies on symmetrically dividing bacteria. In many α-proteobacteria, asymmetric division is a normal part of the life cycle, with each division producing daughter cells with different sizes and fates. Here, we demonstrate that the functionally distinct swarmer and stalked daughter cells produced by the model α-proteobacterium Caulobacter crescentus can have different average growth rates under nutrient-replete conditions despite sharing an identical genome and environment. The discrepancy in growth rate is due to a growth slowdown associated with the cell cycle stage preceding DNA replication (the G1 phase), which initiates in the late predivisional mother cell before daughter cell separation. Both progenies experience a G1-associated growth slowdown, but the effect is more severe in swarmer cells because they have a longer G1 phase. Activity of SpoT, which produces the (p)ppGpp alarmone and extends the G1 phase, accentuates the cell cycle-dependent growth slowdown. Collectively, our data identify a coupling between cell growth, the G1 phase, and asymmetric division that C. crescentus may exploit for environmental adaptation through SpoT activity. This coupling differentially modulates the growth rate of functionally distinct daughter cells, thereby altering the relative abundance of ecologically important G1-specific traits within the population.
Collapse
Affiliation(s)
- Skye Glenn
- Department of Biology, Stanford University, Stanford, CA94305
- Sarafan Chemistry, Engineering, and Medicine for Human Health Institute, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Alessio Fragasso
- Department of Biology, Stanford University, Stanford, CA94305
- Sarafan Chemistry, Engineering, and Medicine for Human Health Institute, Stanford University, Stanford, CA94305
| | - Wei-Hsiang Lin
- Sarafan Chemistry, Engineering, and Medicine for Human Health Institute, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Alexandros Papagiannakis
- Sarafan Chemistry, Engineering, and Medicine for Human Health Institute, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Setsu Kato
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT06511
| | - Christine Jacobs-Wagner
- Department of Biology, Stanford University, Stanford, CA94305
- Sarafan Chemistry, Engineering, and Medicine for Human Health Institute, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
7
|
Chadha Y, Khurana A, Schmoller KM. Eukaryotic cell size regulation and its implications for cellular function and dysfunction. Physiol Rev 2024; 104:1679-1717. [PMID: 38900644 PMCID: PMC11495193 DOI: 10.1152/physrev.00046.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/24/2024] [Accepted: 06/19/2024] [Indexed: 06/22/2024] Open
Abstract
Depending on cell type, environmental inputs, and disease, the cells in the human body can have widely different sizes. In recent years, it has become clear that cell size is a major regulator of cell function. However, we are only beginning to understand how the optimization of cell function determines a given cell's optimal size. Here, we review currently known size control strategies of eukaryotic cells and the intricate link of cell size to intracellular biomolecular scaling, organelle homeostasis, and cell cycle progression. We detail the cell size-dependent regulation of early development and the impact of cell size on cell differentiation. Given the importance of cell size for normal cellular physiology, cell size control must account for changing environmental conditions. We describe how cells sense environmental stimuli, such as nutrient availability, and accordingly adapt their size by regulating cell growth and cell cycle progression. Moreover, we discuss the correlation of pathological states with misregulation of cell size and how for a long time this was considered a downstream consequence of cellular dysfunction. We review newer studies that reveal a reversed causality, with misregulated cell size leading to pathophysiological phenotypes such as senescence and aging. In summary, we highlight the important roles of cell size in cellular function and dysfunction, which could have major implications for both diagnostics and treatment in the clinic.
Collapse
Affiliation(s)
- Yagya Chadha
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Arohi Khurana
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
8
|
Wu W, Lam AR, Suarez K, Smith GN, Duquette SM, Yu J, Mankus D, Bisher M, Lytton-Jean A, Manalis SR, Miettinen TP. Constant surface area-to-volume ratio during cell growth as a design principle in mammalian cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601447. [PMID: 39005340 PMCID: PMC11244959 DOI: 10.1101/2024.07.02.601447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
All cells are subject to geometric constraints, such as surface area-to-volume (SA/V) ratio, that impact cell functions and force biological adaptations. Like the SA/V ratio of a sphere, it is generally assumed that the SA/V ratio of cells decreases as cell size increases. Here, we investigate this in near-spherical mammalian cells using single-cell measurements of cell mass and surface proteins, as well as imaging of plasma membrane morphology. We find that the SA/V ratio remains surprisingly constant as cells grow larger. This observation is largely independent of the cell cycle and the amount of cell growth. Consequently, cell growth results in increased plasma membrane folding, which simplifies cellular design by ensuring sufficient membrane area for cell division, nutrient uptake and deformation at all cell sizes.
Collapse
Affiliation(s)
- Weida Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alice R. Lam
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kayla Suarez
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Grace N. Smith
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sarah M. Duquette
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jiaquan Yu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David Mankus
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Margaret Bisher
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Abigail Lytton-Jean
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Scott R. Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Teemu P. Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
9
|
Miettinen TP, Gomez AL, Wu Y, Wu W, Usherwood TR, Hwang Y, Roller BRK, Polz MF, Manalis SR. Cell size, density, and nutrient dependency of unicellular algal gravitational sinking velocities. SCIENCE ADVANCES 2024; 10:eadn8356. [PMID: 38968348 PMCID: PMC11225777 DOI: 10.1126/sciadv.adn8356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/04/2024] [Indexed: 07/07/2024]
Abstract
Eukaryotic phytoplankton, also known as algae, form the basis of marine food webs and drive marine carbon sequestration. Algae must regulate their motility and gravitational sinking to balance access to light at the surface and nutrients in deeper layers. However, the regulation of gravitational sinking remains largely unknown, especially in motile species. Here, we quantify gravitational sinking velocities according to Stokes' law in diverse clades of unicellular marine microalgae to reveal the cell size, density, and nutrient dependency of sinking velocities. We identify a motile algal species, Tetraselmis sp., that sinks faster when starved due to a photosynthesis-driven accumulation of carbohydrates and a loss of intracellular water, both of which increase cell density. Moreover, the regulation of cell sinking velocities is connected to proliferation and can respond to multiple nutrients. Overall, our work elucidates how cell size and density respond to environmental conditions to drive the vertical migration of motile algae.
Collapse
Affiliation(s)
- Teemu P. Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Annika L. Gomez
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yanqi Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Weida Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Thomas R. Usherwood
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard-MIT Department of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yejin Hwang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Benjamin R. K. Roller
- Division of Microbial Ecology, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, 1030, Austria
| | - Martin F. Polz
- Division of Microbial Ecology, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, 1030, Austria
| | - Scott R. Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
10
|
Scepanovic G, Fernandez-Gonzalez R. Should I shrink or should I grow: cell size changes in tissue morphogenesis. Genome 2024; 67:125-138. [PMID: 38198661 DOI: 10.1139/gen-2023-0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Cells change shape, move, divide, and die to sculpt tissues. Common to all these cell behaviours are cell size changes, which have recently emerged as key contributors to tissue morphogenesis. Cells can change their mass-the number of macromolecules they contain-or their volume-the space they encompass. Changes in cell mass and volume occur through different molecular mechanisms and at different timescales, slow for changes in mass and rapid for changes in volume. Therefore, changes in cell mass and cell volume, which are often linked, contribute to the development and shaping of tissues in different ways. Here, we review the molecular mechanisms by which cells can control and alter their size, and we discuss how changes in cell mass and volume contribute to tissue morphogenesis. The role that cell size control plays in developing embryos is only starting to be elucidated. Research on the signals that control cell size will illuminate our understanding of the cellular and molecular mechanisms that drive tissue morphogenesis.
Collapse
Affiliation(s)
- Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| |
Collapse
|
11
|
Wu W, Ishamuddin SH, Quinn TW, Yerrum S, Zhang Y, Debaize LL, Kao PL, Duquette SM, Murakami MA, Mohseni M, Chow KH, Miettinen TP, Ligon KL, Manalis SR. Measuring single-cell density with high throughput enables dynamic profiling of immune cell and drug response from patient samples. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591092. [PMID: 38712225 PMCID: PMC11071500 DOI: 10.1101/2024.04.25.591092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Cell density, the ratio of cell mass to volume, is an indicator of molecular crowding and therefore a fundamental determinant of cell state and function. However, existing density measurements lack the precision or throughput to quantify subtle differences in cell states, particularly in primary samples. Here we present an approach for measuring the density of 30,000 single cells per hour with a precision of 0.03% (0.0003 g/mL) by integrating fluorescence exclusion microscopy with a suspended microchannel resonator. Applying this approach to human lymphocytes, we discovered that cell density and its variation decrease as cells transition from quiescence to a proliferative state, suggesting that the level of molecular crowding decreases and becomes more regulated upon entry into the cell cycle. Using a pancreatic cancer patient-derived xenograft model, we found that the ex vivo density response of primary tumor cells to drug treatment can predict in vivo tumor growth response. Our method reveals unexpected behavior in molecular crowding during cell state transitions and suggests density as a new biomarker for functional precision medicine.
Collapse
Affiliation(s)
- Weida Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St building 76, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames St #56-651, Cambridge, MA 02139, USA
| | - Sarah H. Ishamuddin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St building 76, Cambridge, MA 02139, USA
| | - Thomas W. Quinn
- Center for Patient-Derived Models, Dana-Farber Cancer Institute, 21 Burlington Ave, Boston, MA 02215, USA
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Smitha Yerrum
- Center for Patient-Derived Models, Dana-Farber Cancer Institute, 21 Burlington Ave, Boston, MA 02215, USA
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Ye Zhang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St building 76, Cambridge, MA 02139, USA
| | - Lydie L. Debaize
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Pei-Lun Kao
- Center for Patient-Derived Models, Dana-Farber Cancer Institute, 21 Burlington Ave, Boston, MA 02215, USA
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Sarah Marie Duquette
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St building 76, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames St #56-651, Cambridge, MA 02139, USA
| | - Mark A. Murakami
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Morvarid Mohseni
- Oncology Discovery, Bristol-Myers Squibb, 250 Water St, Cambridge, MA 02141, USA
| | - Kin-Hoe Chow
- Center for Patient-Derived Models, Dana-Farber Cancer Institute, 21 Burlington Ave, Boston, MA 02215, USA
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Teemu P. Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St building 76, Cambridge, MA 02139, USA
| | - Keith L. Ligon
- Center for Patient-Derived Models, Dana-Farber Cancer Institute, 21 Burlington Ave, Boston, MA 02215, USA
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
- Broad Institute of Harvard and MIT, 415 Main St, Cambridge, MA 02142, USA
- Department of Pathology, Brigham & Women’s Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02215, USA
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Scott R. Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main St building 76, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames St #56-651, Cambridge, MA 02139, USA
- Broad Institute of Harvard and MIT, 415 Main St, Cambridge, MA 02142, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 33 Massachusetts Ave, Cambridge, MA 02139, USA
| |
Collapse
|
12
|
Pinto SC, Stojilković B, Zhang X, Sablowski R. Plant cell size: Links to cell cycle, differentiation and ploidy. CURRENT OPINION IN PLANT BIOLOGY 2024; 78:102527. [PMID: 38484440 DOI: 10.1016/j.pbi.2024.102527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 04/07/2024]
Abstract
Cell size affects many processes, including exchange of nutrients and external signals, cell division and tissue mechanics. Across eukaryotes, cells have evolved mechanisms that assess their own size to inform processes such as cell cycle progression or gene expression. Here, we review recent progress in understanding plant cell size regulation and its implications, relating these findings to work in other eukaryotes. Highlights include use of DNA contents as reference point to control the cell cycle in shoot meristems, a size-dependent cell fate decision during stomatal development and insights into the interconnection between ploidy, cell size and cell wall mechanics.
Collapse
Affiliation(s)
- Sara C Pinto
- Cell and Developmental Biology, John Innes Centre, Norwich, UK
| | | | - Xinyu Zhang
- Cell and Developmental Biology, John Innes Centre, Norwich, UK
| | | |
Collapse
|
13
|
Lessenger AT, Swaffer MP, Skotheim JM, Feldman JL. Somatic polyploidy supports biosynthesis and tissue function by increasing transcriptional output. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.586714. [PMID: 38585999 PMCID: PMC10996643 DOI: 10.1101/2024.03.25.586714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Cell size and biosynthetic capacity generally increase with increased DNA content. Polyploidy has therefore been proposed to be an adaptive strategy to increase cell size in specialized tissues with high biosynthetic demands. However, if and how DNA concentration limits cellular biosynthesis in vivo is not well understood, and the impacts of polyploidy in non-disease states is not well studied. Here, we show that polyploidy in the C. elegans intestine is critical for cell growth and yolk biosynthesis, a central role of this organ. Artificially lowering the DNA/cytoplasm ratio by reducing polyploidization in the intestine gave rise to smaller cells with more dilute mRNA. Highly-expressed transcripts were more sensitive to this mRNA dilution, whereas lowly-expressed genes were partially compensated - in part by loading more RNA Polymerase II on the remaining genomes. DNA-dilute cells had normal total protein concentration, which we propose is achieved by increasing production of translational machinery at the expense of specialized, cell-type specific proteins.
Collapse
|
14
|
Diehl FF, Sapp KM, Vander Heiden MG. The bidirectional relationship between metabolism and cell cycle control. Trends Cell Biol 2024; 34:136-149. [PMID: 37385879 DOI: 10.1016/j.tcb.2023.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023]
Abstract
The relationship between metabolism and cell cycle progression is complex and bidirectional. Cells must rewire metabolism to meet changing biosynthetic demands across cell cycle phases. In turn, metabolism can influence cell cycle progression through direct regulation of cell cycle proteins, through nutrient-sensing signaling pathways, and through its impact on cell growth, which is linked to cell division. Furthermore, metabolism is a key player in mediating quiescence-proliferation transitions in physiologically important cell types, such as stem cells. How metabolism impacts cell cycle progression, exit, and re-entry, as well as how these processes impact metabolism, is not fully understood. Recent advances uncovering mechanistic links between cell cycle regulators and metabolic processes demonstrate a complex relationship between metabolism and cell cycle control, with many questions remaining.
Collapse
Affiliation(s)
- Frances F Diehl
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kiera M Sapp
- Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
15
|
Liu X, Yan J, Kirschner MW. Cell size homeostasis is tightly controlled throughout the cell cycle. PLoS Biol 2024; 22:e3002453. [PMID: 38180950 PMCID: PMC10769027 DOI: 10.1371/journal.pbio.3002453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 01/07/2024] Open
Abstract
To achieve a stable size distribution over multiple generations, proliferating cells require a means of counteracting stochastic noise in the rate of growth, the time spent in various phases of the cell cycle, and the imprecision in the placement of the plane of cell division. In the most widely accepted model, cell size is thought to be regulated at the G1/S transition, such that cells smaller than a critical size pause at the end of G1 phase until they have accumulated mass to a predetermined size threshold, at which point the cells proceed through the rest of the cell cycle. However, a model, based solely on a specific size checkpoint at G1/S, cannot readily explain why cells with deficient G1/S control mechanisms are still able to maintain a very stable cell size distribution. Furthermore, such a model would not easily account for stochastic variation in cell size during the subsequent phases of the cell cycle, which cannot be anticipated at G1/S. To address such questions, we applied computationally enhanced quantitative phase microscopy (ceQPM) to populations of cultured human cell lines, which enables highly accurate measurement of cell dry mass of individual cells throughout the cell cycle. From these measurements, we have evaluated the factors that contribute to maintaining cell mass homeostasis at any point in the cell cycle. Our findings reveal that cell mass homeostasis is accurately maintained, despite disruptions to the normal G1/S machinery or perturbations in the rate of cell growth. Control of cell mass is generally not confined to regulation of the G1 length. Instead mass homeostasis is imposed throughout the cell cycle. In the cell lines examined, we find that the coefficient of variation (CV) in dry mass of cells in the population begins to decline well before the G1/S transition and continues to decline throughout S and G2 phases. Among the different cell types tested, the detailed response of cell growth rate to cell mass differs. However, in general, when it falls below that for exponential growth, the natural increase in the CV of cell mass is effectively constrained. We find that both mass-dependent cell cycle regulation and mass-dependent growth rate modulation contribute to reducing cell mass variation within the population. Through the interplay and coordination of these 2 processes, accurate cell mass homeostasis emerges. Such findings reveal previously unappreciated and very general principles of cell size control in proliferating cells. These same regulatory processes might also be operative in terminally differentiated cells. Further quantitative dynamical studies should lead to a better understanding of the underlying molecular mechanisms of cell size control.
Collapse
Affiliation(s)
- Xili Liu
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jiawei Yan
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Marc W. Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
16
|
Golding I, Amir A. Gene expression in growing cells: A biophysical primer. ARXIV 2023:arXiv:2311.12143v1. [PMID: 38045483 PMCID: PMC10690283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Cell growth and gene expression, two essential elements of all living systems, have long been the focus of biophysical interrogation. Advances in experimental single-cell methods have invigorated theoretical studies into these processes. However, until recently, there was little dialog between the two areas of study. In particular, most theoretical models for gene regulation assumed gene activity to be oblivious to the progression of the cell cycle between birth and division. But, in fact, there are numerous ways in which the periodic character of all cellular observables can modulate gene expression. The molecular factors required for transcription and translation-RNA polymerase, transcription factors, ribosomes-increase in number during the cell cycle, but are also diluted due to the continuous increase in cell volume. The replication of the genome changes the dosage of those same cellular players but also provides competing targets for regulatory binding. Finally, cell division reduces their number again, and so forth. Stochasticity is inherent to all these biological processes, manifested in fluctuations in the synthesis and degradation of new cellular components as well as the random partitioning of molecules at each cell division event. The notion of gene expression as stationary is thus hard to justify. In this review, we survey the emerging paradigm of cell-cycle regulated gene expression, with an emphasis on the global expression patterns rather than gene-specific regulation. We discuss recent experimental reports where cell growth and gene expression were simultaneously measured in individual cells, providing first glimpses into the coupling between the two, and motivating several questions. How do the levels of gene expression products - mRNA and protein - scale with the cell volume and cell-cycle progression? What are the molecular origins of the observed scaling laws, and when do they break down to yield non-canonical behavior? What are the consequences of cell-cycle dependence for the heterogeneity ("noise") in gene expression within a cell population? While the experimental findings, not surprisingly, differ among genes, organisms, and environmental conditions, several theoretical models have emerged that attempt to reconcile these differences and form a unifying framework for understanding gene expression in growing cells.
Collapse
Affiliation(s)
- Ido Golding
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ariel Amir
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
17
|
Lanz MC, Zhang S, Swaffer MP, Hernández Götz L, McCarty F, Ziv I, Jarosz DF, Elias JE, Skotheim JM. Genome dilution by cell growth drives starvation-like proteome remodeling in mammalian and yeast cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562558. [PMID: 37905015 PMCID: PMC10614910 DOI: 10.1101/2023.10.16.562558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cell size is tightly controlled in healthy tissues and single-celled organisms, but it remains unclear how size influences cell physiology. Increasing cell size was recently shown to remodel the proteomes of cultured human cells, demonstrating that large and small cells of the same type can be biochemically different. Here, we corroborate these results in mouse hepatocytes and extend our analysis using yeast. We find that size-dependent proteome changes are highly conserved and mostly independent of metabolic state. As eukaryotic cells grow larger, the dilution of the genome elicits a starvation-like proteome phenotype, suggesting that growth in large cells is limited by the genome in a manner analogous to a limiting nutrient. We also demonstrate that the proteomes of replicatively-aged yeast are primarily determined by their large size. Overall, our data suggest that genome concentration is a universal determinant of proteome content in growing cells.
Collapse
|
18
|
Hatton IA, Galbraith ED, Merleau NSC, Miettinen TP, Smith BM, Shander JA. The human cell count and size distribution. Proc Natl Acad Sci U S A 2023; 120:e2303077120. [PMID: 37722043 PMCID: PMC10523466 DOI: 10.1073/pnas.2303077120] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/24/2023] [Indexed: 09/20/2023] Open
Abstract
Cell size and cell count are adaptively regulated and intimately linked to growth and function. Yet, despite their widespread relevance, the relation between cell size and count has never been formally examined over the whole human body. Here, we compile a comprehensive dataset of cell size and count over all major cell types, with data drawn from >1,500 published sources. We consider the body of a representative male (70 kg), which allows further estimates of a female (60 kg) and 10-y-old child (32 kg). We build a hierarchical interface for the cellular organization of the body, giving easy access to data, methods, and sources (https://humancelltreemap.mis.mpg.de/). In total, we estimate total body counts of ≈36 trillion cells in the male, ≈28 trillion in the female, and ≈17 trillion in the child. These data reveal a surprising inverse relation between cell size and count, implying a trade-off between these variables, such that all cells within a given logarithmic size class contribute an equal fraction to the body's total cellular biomass. We also find that the coefficient of variation is approximately independent of mean cell size, implying the existence of cell-size regulation across cell types. Our data serve to establish a holistic quantitative framework for the cells of the human body, and highlight large-scale patterns in cell biology.
Collapse
Affiliation(s)
- Ian A. Hatton
- Max Planck Institute for Mathematics in the Sciences, Leipzig04103, Germany
- Department of Earth and Planetary Sciences, McGill University, Montreal, QuebecH3A 0E8, Canada
| | - Eric D. Galbraith
- Department of Earth and Planetary Sciences, McGill University, Montreal, QuebecH3A 0E8, Canada
- ICREA, Barcelona08010, Spain
| | - Nono S. C. Merleau
- Max Planck Institute for Mathematics in the Sciences, Leipzig04103, Germany
- Center for Scalable Data Analytics and Artificial Intelligence, University of Leipzig, D-04105Leipzig, Germany
| | - Teemu P. Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Benjamin McDonald Smith
- Department of Medicine, McGill University Health Centre Research Institute, Montreal, QuebecH4A 3S5, Canada
- Department of Medicine, Columbia University Medical Center, New York, NY10032
| | | |
Collapse
|
19
|
Charan MR, Augustsson P. Acoustophoretic Characterization and Separation of Blood Cells in Acoustic Impedance Gradients. PHYSICAL REVIEW APPLIED 2023; 20:024066. [PMID: 38333566 PMCID: PMC7615610 DOI: 10.1103/physrevapplied.20.024066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Single-cell phenotyping based on biophysical properties is a promising tool to distinguish cell types and their response to a given condition, and charting such properties also enables optimization of cell separations. Isoacoustic focusing, where cells migrate to their points of zero acoustic contrast in an acoustic impedance gradient, added the effective acoustic impedance of cells to the directory of biophysical properties that can be utilized to categorize or separate cells. This study investigates isoacoustic focusing in a stop-flow regime and shows how cells migrate towards their isoacoustic point. We introduce a numerical model that we use to estimate the acoustic energy density in acoustic impedance gradient media by tracking particles of known properties, and we investigate the effect of acoustic streaming. From the measured trajectories of cells combined with fluorescence intensity images of the slowly diffusing gradient, we read out the effective acoustic impedance of neutrophils and K562 cancer cells. Finally, we propose suitable acoustic impedance gradients that lead to a high degree separation of neutrophils and K562 cells in a continuous-flow configuration.
Collapse
Affiliation(s)
- Mahdi Rezayati Charan
- Department of Biomedical Engineering, Lund University, Ole Römers Väg 3, 22363 Lund, Sweden
| | - Per Augustsson
- Department of Biomedical Engineering, Lund University, Ole Römers Väg 3, 22363 Lund, Sweden
| |
Collapse
|
20
|
Jiang N, Malone M, Chizari S. Antigen-specific and cross-reactive T cells in protection and disease. Immunol Rev 2023; 316:120-135. [PMID: 37209375 PMCID: PMC10524458 DOI: 10.1111/imr.13217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/22/2023]
Abstract
Human T cells have a diverse T-cell receptor (TCR) repertoire that endows them with the ability to identify and defend against a broad spectrum of antigens. The universe of possible antigens that T cells may encounter, however, is even larger. To effectively surveil such a vast universe, the T-cell repertoire must adopt a high degree of cross-reactivity. Likewise, antigen-specific and cross-reactive T-cell responses play pivotal roles in both protective and pathological immune responses in numerous diseases. In this review, we explore the implications of these antigen-driven T-cell responses, with a particular focus on CD8+ T cells, using infection, neurodegeneration, and cancer as examples. We also summarize recent technological advances that facilitate high-throughput profiling of antigen-specific and cross-reactive T-cell responses experimentally, as well as computational biology approaches that predict these interactions.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, 19104
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, 19104
| | - Michael Malone
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| | - Shahab Chizari
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
21
|
Fung HF, Bergmann DC. Function follows form: How cell size is harnessed for developmental decisions. Eur J Cell Biol 2023; 102:151312. [PMID: 36989838 DOI: 10.1016/j.ejcb.2023.151312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Cell size has profound effects on biological function, influencing a wide range of processes, including biosynthetic capacity, metabolism, and nutrient uptake. As a result, size is typically maintained within a narrow, population-specific range through size control mechanisms, which are an active area of study. While the physiological consequences of cell size are relatively well-characterized, less is known about its developmental consequences, and specifically its effects on developmental transitions. In this review, we compare systems where cell size is linked to developmental transitions, paying particular attention to examples from plants. We conclude by proposing that size can offer a simple readout of complex inputs, enabling flexible decisions during plant development.
Collapse
|
22
|
Diaz-Cuadros M, Miettinen TP, Skinner OS, Sheedy D, Díaz-García CM, Gapon S, Hubaud A, Yellen G, Manalis SR, Oldham WM, Pourquié O. Metabolic regulation of species-specific developmental rates. Nature 2023; 613:550-557. [PMID: 36599986 PMCID: PMC9944513 DOI: 10.1038/s41586-022-05574-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/18/2022] [Indexed: 01/06/2023]
Abstract
Animals display substantial inter-species variation in the rate of embryonic development despite a broad conservation of the overall sequence of developmental events. Differences in biochemical reaction rates, including the rates of protein production and degradation, are thought to be responsible for species-specific rates of development1-3. However, the cause of differential biochemical reaction rates between species remains unknown. Here, using pluripotent stem cells, we have established an in vitro system that recapitulates the twofold difference in developmental rate between mouse and human embryos. This system provides a quantitative measure of developmental speed as revealed by the period of the segmentation clock, a molecular oscillator associated with the rhythmic production of vertebral precursors. Using this system, we show that mass-specific metabolic rates scale with the developmental rate and are therefore higher in mouse cells than in human cells. Reducing these metabolic rates by inhibiting the electron transport chain slowed down the segmentation clock by impairing the cellular NAD+/NADH redox balance and, further downstream, lowering the global rate of protein synthesis. Conversely, increasing the NAD+/NADH ratio in human cells by overexpression of the Lactobacillus brevis NADH oxidase LbNOX increased the translation rate and accelerated the segmentation clock. These findings represent a starting point for the manipulation of developmental rate, with multiple translational applications including accelerating the differentiation of human pluripotent stem cells for disease modelling and cell-based therapies.
Collapse
Affiliation(s)
- Margarete Diaz-Cuadros
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
| | - Teemu P Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Owen S Skinner
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Dylan Sheedy
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Carlos Manlio Díaz-García
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Svetlana Gapon
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Alexis Hubaud
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William M Oldham
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
23
|
Davies DM, van den Handel K, Bharadwaj S, Lengefeld J. Cellular enlargement - A new hallmark of aging? Front Cell Dev Biol 2022; 10:1036602. [PMID: 36438561 PMCID: PMC9688412 DOI: 10.3389/fcell.2022.1036602] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2023] Open
Abstract
Years of important research has revealed that cells heavily invest in regulating their size. Nevertheless, it has remained unclear why accurate size control is so important. Our recent study using hematopoietic stem cells (HSCs) in vivo indicates that cellular enlargement is causally associated with aging. Here, we present an overview of these findings and their implications. Furthermore, we performed a broad literature analysis to evaluate the potential of cellular enlargement as a new aging hallmark and to examine its connection to previously described aging hallmarks. Finally, we highlight interesting work presenting a correlation between cell size and age-related diseases. Taken together, we found mounting evidence linking cellular enlargement to aging and age-related diseases. Therefore, we encourage researchers from seemingly unrelated areas to take a fresh look at their data from the perspective of cell size.
Collapse
Affiliation(s)
- Daniel M. Davies
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kim van den Handel
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Soham Bharadwaj
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jette Lengefeld
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Liu S, Tan C, Tyers M, Zetterberg A, Kafri R. What programs the size of animal cells? Front Cell Dev Biol 2022; 10:949382. [PMID: 36393871 PMCID: PMC9665425 DOI: 10.3389/fcell.2022.949382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/07/2022] [Indexed: 01/19/2023] Open
Abstract
The human body is programmed with definite quantities, magnitudes, and proportions. At the microscopic level, such definite sizes manifest in individual cells - different cell types are characterized by distinct cell sizes whereas cells of the same type are highly uniform in size. How do cells in a population maintain uniformity in cell size, and how are changes in target size programmed? A convergence of recent and historical studies suggest - just as a thermostat maintains room temperature - the size of proliferating animal cells is similarly maintained by homeostatic mechanisms. In this review, we first summarize old and new literature on the existence of cell size checkpoints, then discuss additional advances in the study of size homeostasis that involve feedback regulation of cellular growth rate. We further discuss recent progress on the molecules that underlie cell size checkpoints and mechanisms that specify target size setpoints. Lastly, we discuss a less-well explored teleological question: why does cell size matter and what is the functional importance of cell size control?
Collapse
Affiliation(s)
- Shixuan Liu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, United States
| | - Ceryl Tan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, University of Montréal, Montréal, QC, Canada
| | - Anders Zetterberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ran Kafri
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
25
|
Øvrebø JI, Ma Y, Edgar BA. Cell growth and the cell cycle: New insights about persistent questions. Bioessays 2022; 44:e2200150. [PMID: 36222263 DOI: 10.1002/bies.202200150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/08/2022]
Abstract
Before a cell divides into two daughter cells, it typically doubles not only its DNA, but also its mass. Numerous studies in cells ranging from yeast to mammals have shown that cellular growth, stimulated by nutrients and/or growth factor signaling, is a prerequisite for cell cycle progression in most types of cells. The textbook view of growth-regulated cell cycles is that growth signaling activates the transcription of G1 Cyclin genes to induce cell proliferation, and also stimulates anabolic metabolism and cell growth in parallel. However, genetic knockout tests in model organisms indicate that this is not the whole story, and new studies show that additional, "smarter" mechanisms help to coordinate the cell cycle with growth itself. Here we summarize recent advances in this field, and discuss current models in which growth signaling regulates cell proliferation by targeting core cell cycle regulators via non-transcriptional mechanisms.
Collapse
Affiliation(s)
- Jan Inge Øvrebø
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Yiqin Ma
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Bruce A Edgar
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
26
|
Chatzitheodoridou D, D'Ario M, Jones I, Piñeros L, Serbanescu D, O'Donnell F, Cadart C, Swaffer MP. Meeting report - Cell size and growth: from single cells to the tree of life. J Cell Sci 2022; 135:jcs260634. [PMID: 36259425 DOI: 10.1242/jcs.260634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023] Open
Abstract
In April 2022, The Company of Biologists hosted their first post-pandemic in-person Workshop at Buxted Park Country House in the Sussex countryside. The Workshop, entitled 'Cell size and growth: from single cells to the tree of life', gathered a small group of early-career and senior researchers with expertise in cell size spanning a broad range of organisms, including bacteria, yeast, animal cells, embryos and plants, and working in fields from cell biology to ecology and evolutionary biology. The programme made ample room for fruitful discussions and provided a much-needed opportunity to discuss the most recent findings relating to the regulation of cell size and growth, identify the emerging challenges for the field, and build a community after the pandemic.
Collapse
Affiliation(s)
| | - Marco D'Ario
- Department of Cell and Developmental Biology, John Innes Centre, Norwich, NR4 7UH, UK
| | - Ian Jones
- Department of Cancer Biology, Chester Beatty Laboratories, Institute of Cancer Research, London, SW3 6JB, UK
| | - Liliana Piñeros
- Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O&N1bis gebouw 402-20, Herestraat 49, B-3000 Leuven, Belgium
| | - Diana Serbanescu
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London, WC1E 6BT, UK
| | - Frank O'Donnell
- The Company of Biologists, 94 Station Road, Histon, Cambridge, CB24 9LF, UK
| | - Clotilde Cadart
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | | |
Collapse
|
27
|
Ko J, Khan F, Nam Y, Lee BJ, Lee J. Nanomechanical Sensing Using Heater-Integrated Fluidic Resonators. NANO LETTERS 2022; 22:7768-7775. [PMID: 35980246 DOI: 10.1021/acs.nanolett.2c01572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Micro/nanochannel resonators have been used to measure cells, suspended nanoparticles, or liquids, primarily at or near room temperature while their high temperature operation can offer promising applications such as calorimetric measurements and thermogravimetric analysis. To date, global electrothermal or local photothermal heating mechanisms have been attempted for channel resonators, but both approaches are intrinsically limited by a narrow temperature modulation range, slow heating/cooling, less quantitative heating, or time-consuming optical alignment. Here, we introduce heater-integrated fluidic resonators (HFRs) that enable fast, quantitative, alignment-free, and wide-range temperature modulation and simultaneously offer resistive thermometry and resonant densitometry. HFRs with or without a dispensing nozzle are fabricated, thoroughly characterized, and used for high throughput thermophysical properties measurements, microchannel boiling studies, and atomized spray dispensing. The HFR, without a doubt, opens a new avenue for nanoscale thermal analysis and processing and further encourages the integration of additional functions into channel resonators.
Collapse
Affiliation(s)
- Juhee Ko
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
- Center for Extreme Thermal Physics and Manufacturing, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Faheem Khan
- Life Analytical Inc., Edmonton, Alberta T6B 2N2, Canada
| | - Youngsuk Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
- Center for Extreme Thermal Physics and Manufacturing, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Bong Jae Lee
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
- Center for Extreme Thermal Physics and Manufacturing, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Jungchul Lee
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
- Center for Extreme Thermal Physics and Manufacturing, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| |
Collapse
|
28
|
Abstract
The most fundamental feature of cellular form is size, which sets the scale of all cell biological processes. Growth, form, and function are all necessarily linked in cell biology, but we often do not understand the underlying molecular mechanisms nor their specific functions. Here, we review progress toward determining the molecular mechanisms that regulate cell size in yeast, animals, and plants, as well as progress toward understanding the function of cell size regulation. It has become increasingly clear that the mechanism of cell size regulation is deeply intertwined with basic mechanisms of biosynthesis, and how biosynthesis can be scaled (or not) in proportion to cell size. Finally, we highlight recent findings causally linking aberrant cell size regulation to cellular senescence and their implications for cancer therapies.
Collapse
Affiliation(s)
- Shicong Xie
- Department of Biology, Stanford University, Stanford, California, USA;
| | - Matthew Swaffer
- Department of Biology, Stanford University, Stanford, California, USA;
| | - Jan M Skotheim
- Department of Biology, Stanford University, Stanford, California, USA;
- Chan Zuckerberg Biohub, San Francisco, California, USA
| |
Collapse
|
29
|
Doan-Nguyen TP, Crespy D. Advanced density-based methods for the characterization of materials, binding events, and kinetics. Chem Soc Rev 2022; 51:8612-8651. [PMID: 36172819 DOI: 10.1039/d1cs00232e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Investigations of the densities of chemicals and materials bring valuable insights into the fundamental understanding of matter and processes. Recently, advanced density-based methods have been developed with wide measurement ranges (i.e. 0-23 g cm-3), high resolutions (i.e. 10-6 g cm-3), compatibility with different types of samples and the requirement of extremely low volumes of sample (as low as a single cell). Certain methods, such as magnetic levitation, are inexpensive, portable and user-friendly. Advanced density-based methods are, therefore, beneficially used to obtain absolute density values, composition of mixtures, characteristics of binding events, and kinetics of chemical and biological processes. Herein, the principles and applications of magnetic levitation, acoustic levitation, electrodynamic balance, aqueous multiphase systems, and suspended microchannel resonators for materials science are discussed.
Collapse
Affiliation(s)
- Thao P Doan-Nguyen
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand. .,Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Daniel Crespy
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand. .,Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| |
Collapse
|
30
|
Zatulovskiy E, Lanz MC, Zhang S, McCarthy F, Elias JE, Skotheim JM. Delineation of proteome changes driven by cell size and growth rate. Front Cell Dev Biol 2022; 10:980721. [PMID: 36133920 PMCID: PMC9483106 DOI: 10.3389/fcell.2022.980721] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/09/2022] [Indexed: 01/10/2023] Open
Abstract
Increasing cell size drives changes to the proteome, which affects cell physiology. As cell size increases, some proteins become more concentrated while others are diluted. As a result, the state of the cell changes continuously with increasing size. In addition to these proteomic changes, large cells have a lower growth rate (protein synthesis rate per unit volume). That both the cell's proteome and growth rate change with cell size suggests they may be interdependent. To test this, we used quantitative mass spectrometry to measure how the proteome changes in response to the mTOR inhibitor rapamycin, which decreases the cellular growth rate and has only a minimal effect on cell size. We found that large cell size and mTOR inhibition, both of which lower the growth rate of a cell, remodel the proteome in similar ways. This suggests that many of the effects of cell size are mediated by the size-dependent slowdown of the cellular growth rate. For example, the previously reported size-dependent expression of some senescence markers could reflect a cell's declining growth rate rather than its size per se. In contrast, histones and other chromatin components are diluted in large cells independently of the growth rate, likely so that they remain in proportion with the genome. Finally, size-dependent changes to the cell's growth rate and proteome composition are still apparent in cells continually exposed to a saturating dose of rapamycin, which indicates that cell size can affect the proteome independently of mTORC1 signaling. Taken together, our results clarify the dependencies between cell size, growth, mTOR activity, and the proteome remodeling that ultimately controls many aspects of cell physiology.
Collapse
Affiliation(s)
| | - Michael C. Lanz
- Department of Biology, Stanford University, Stanford, CA, United States
- Chan Zuckerberg Biohub, Stanford, CA, United States
| | - Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA, United States
| | | | | | - Jan M. Skotheim
- Department of Biology, Stanford University, Stanford, CA, United States
- Chan Zuckerberg Biohub, Stanford, CA, United States
| |
Collapse
|
31
|
Lanz MC, Zatulovskiy E, Swaffer MP, Zhang L, Ilerten I, Zhang S, You DS, Marinov G, McAlpine P, Elias JE, Skotheim JM. Increasing cell size remodels the proteome and promotes senescence. Mol Cell 2022; 82:3255-3269.e8. [PMID: 35987199 PMCID: PMC9444988 DOI: 10.1016/j.molcel.2022.07.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/06/2022] [Accepted: 07/25/2022] [Indexed: 01/10/2023]
Abstract
Cell size is tightly controlled in healthy tissues, but it is unclear how deviations in cell size affect cell physiology. To address this, we measured how the cell's proteome changes with increasing cell size. Size-dependent protein concentration changes are widespread and predicted by subcellular localization, size-dependent mRNA concentrations, and protein turnover. As proliferating cells grow larger, concentration changes typically associated with cellular senescence are increasingly pronounced, suggesting that large size may be a cause rather than just a consequence of cell senescence. Consistent with this hypothesis, larger cells are prone to replicative, DNA-damage-induced, and CDK4/6i-induced senescence. Size-dependent changes to the proteome, including those associated with senescence, are not observed when an increase in cell size is accompanied by an increase in ploidy. Together, our findings show how cell size could impact many aspects of cell physiology by remodeling the proteome and provide a rationale for cell size control and polyploidization.
Collapse
Affiliation(s)
- Michael C Lanz
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | | | | | | | - Ilayda Ilerten
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Dong Shin You
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Georgi Marinov
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | | | | | - Jan M Skotheim
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
32
|
Zhang S, Zatulovskiy E, Arand J, Sage J, Skotheim JM. The cell cycle inhibitor RB is diluted in G1 and contributes to controlling cell size in the mouse liver. Front Cell Dev Biol 2022; 10:965595. [PMID: 36092730 PMCID: PMC9452963 DOI: 10.3389/fcell.2022.965595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/27/2022] [Indexed: 12/14/2022] Open
Abstract
Every type of cell in an animal maintains a specific size, which likely contributes to its ability to perform its physiological functions. While some cell size control mechanisms are beginning to be elucidated through studies of cultured cells, it is unclear if and how such mechanisms control cell size in an animal. For example, it was recently shown that RB, the retinoblastoma protein, was diluted by cell growth in G1 to promote size-dependence of the G1/S transition. However, it remains unclear to what extent the RB-dilution mechanism controls cell size in an animal. We therefore examined the contribution of RB-dilution to cell size control in the mouse liver. Consistent with the RB-dilution model, genetic perturbations decreasing RB protein concentrations through inducible shRNA expression or through liver-specific Rb1 knockout reduced hepatocyte size, while perturbations increasing RB protein concentrations in an Fah -/- mouse model increased hepatocyte size. Moreover, RB concentration reflects cell size in G1 as it is lower in larger G1 hepatocytes. In contrast, concentrations of the cell cycle activators Cyclin D1 and E2f1 were relatively constant. Lastly, loss of Rb1 weakened cell size control, i.e., reduced the inverse correlation between how much cells grew in G1 and how large they were at birth. Taken together, our results show that an RB-dilution mechanism contributes to cell size control in the mouse liver by linking cell growth to the G1/S transition.
Collapse
Affiliation(s)
- Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA, United States
| | | | - Julia Arand
- Departments of Pediatrics and Genetics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Julien Sage
- Departments of Pediatrics and Genetics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jan M. Skotheim
- Department of Biology, Stanford University, Stanford, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
33
|
Polanco ER, Moustafa TE, Butterfield A, Scherer SD, Cortes-Sanchez E, Bodily T, Spike BT, Welm BE, Bernard PS, Zangle TA. Multiparametric quantitative phase imaging for real-time, single cell, drug screening in breast cancer. Commun Biol 2022; 5:794. [PMID: 35941353 PMCID: PMC9360018 DOI: 10.1038/s42003-022-03759-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 07/22/2022] [Indexed: 11/09/2022] Open
Abstract
Quantitative phase imaging (QPI) measures the growth rate of individual cells by quantifying changes in mass versus time. Here, we use the breast cancer cell lines MCF-7, BT-474, and MDA-MB-231 to validate QPI as a multiparametric approach for determining response to single-agent therapies. Our method allows for rapid determination of drug sensitivity, cytotoxicity, heterogeneity, and time of response for up to 100,000 individual cells or small clusters in a single experiment. We find that QPI EC50 values are concordant with CellTiter-Glo (CTG), a gold standard metabolic endpoint assay. In addition, we apply multiparametric QPI to characterize cytostatic/cytotoxic and rapid/slow responses and track the emergence of resistant subpopulations. Thus, QPI reveals dynamic changes in response heterogeneity in addition to average population responses, a key advantage over endpoint viability or metabolic assays. Overall, multiparametric QPI reveals a rich picture of cell growth by capturing the dynamics of single-cell responses to candidate therapies.
Collapse
Affiliation(s)
- Edward R Polanco
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Tarek E Moustafa
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Andrew Butterfield
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Sandra D Scherer
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Emilio Cortes-Sanchez
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Tyler Bodily
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Benjamin T Spike
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Bryan E Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | - Philip S Bernard
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, UT, USA
| | - Thomas A Zangle
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
34
|
Cadart C, Heald R. Scaling of biosynthesis and metabolism with cell size. Mol Biol Cell 2022; 33:pe5. [PMID: 35862496 PMCID: PMC9582640 DOI: 10.1091/mbc.e21-12-0627] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
Cells adopt a size that is optimal for their function, and pushing them beyond this limit can cause cell aging and death by senescence or reduce proliferative potential. However, by increasing their genome copy number (ploidy), cells can increase their size dramatically and homeostatically maintain physiological properties such as biosynthesis rate. Recent studies investigating the relationship between cell size and rates of biosynthesis and metabolism under normal, polyploid, and pathological conditions are revealing new insights into how cells attain the best function or fitness for their size by tuning processes including transcription, translation, and mitochondrial respiration. A new frontier is to connect single-cell scaling relationships with tissue and whole-organism physiology, which promises to reveal molecular and evolutionary principles underlying the astonishing diversity of size observed across the tree of life.
Collapse
Affiliation(s)
- Clotilde Cadart
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA 94720-3200
| | - Rebecca Heald
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA 94720-3200
| |
Collapse
|
35
|
Diehl FF, Miettinen TP, Elbashir R, Nabel CS, Darnell AM, Do BT, Manalis SR, Lewis CA, Vander Heiden MG. Nucleotide imbalance decouples cell growth from cell proliferation. Nat Cell Biol 2022; 24:1252-1264. [PMID: 35927450 PMCID: PMC9359916 DOI: 10.1038/s41556-022-00965-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 06/21/2022] [Indexed: 12/26/2022]
Abstract
Nucleotide metabolism supports RNA synthesis and DNA replication to enable cell growth and division. Nucleotide depletion can inhibit cell growth and proliferation, but how cells sense and respond to changes in the relative levels of individual nucleotides is unclear. Moreover, the nucleotide requirement for biomass production changes over the course of the cell cycle, and how cells coordinate differential nucleotide demands with cell cycle progression is not well understood. Here we find that excess levels of individual nucleotides can inhibit proliferation by disrupting the relative levels of nucleotide bases needed for DNA replication and impeding DNA replication. The resulting purine and pyrimidine imbalances are not sensed by canonical growth regulatory pathways like mTORC1, Akt and AMPK signalling cascades, causing excessive cell growth despite inhibited proliferation. Instead, cells rely on replication stress signalling to survive during, and recover from, nucleotide imbalance during S phase. We find that ATR-dependent replication stress signalling is activated during unperturbed S phases and promotes nucleotide availability to support DNA replication. Together, these data reveal that imbalanced nucleotide levels are not detected until S phase, rendering cells reliant on replication stress signalling to cope with this metabolic problem and disrupting the coordination of cell growth and division.
Collapse
Affiliation(s)
- Frances F Diehl
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Teemu P Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Ryan Elbashir
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christopher S Nabel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Alicia M Darnell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brian T Do
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, USA
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Departments of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
36
|
Miettinen TP, Ly KS, Lam A, Manalis SR. Single-cell monitoring of dry mass and dry mass density reveals exocytosis of cellular dry contents in mitosis. eLife 2022; 11:e76664. [PMID: 35535854 PMCID: PMC9090323 DOI: 10.7554/elife.76664] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/22/2022] [Indexed: 01/02/2023] Open
Abstract
Cell mass and composition change with cell cycle progression. Our previous work characterized buoyant mass dynamics in mitosis (Miettinen et al., 2019), but how dry mass and cell composition change in mitosis has remained unclear. To better understand mitotic cell growth and compositional changes, we develop a single-cell approach for monitoring dry mass and the density of that dry mass every ~75 s with 1.3% and 0.3% measurement precision, respectively. We find that suspension grown mammalian cells lose dry mass and increase dry mass density following mitotic entry. These changes display large, non-genetic cell-to-cell variability, and the changes are reversed at metaphase-anaphase transition, after which dry mass continues accumulating. The change in dry mass density causes buoyant and dry mass to differ specifically in early mitosis, thus reconciling existing literature on mitotic cell growth. Mechanistically, cells in early mitosis increase lysosomal exocytosis, and inhibition of lysosomal exocytosis decreases the dry mass loss and dry mass density increase in mitosis. Overall, our work provides a new approach for monitoring single-cell dry mass and dry mass density, and reveals that mitosis is coupled to extensive exocytosis-mediated secretion of cellular contents.
Collapse
Affiliation(s)
- Teemu P Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeUnited States
- MIT Center for Precision Cancer Medicine, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Kevin S Ly
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Alice Lam
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeUnited States
- MIT Center for Precision Cancer Medicine, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Mechanical Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
37
|
Li B, Maslan A, Kitayama SE, Pierce C, Streets AM, Sohn LL. Mechanical phenotyping reveals unique biomechanical responses in retinoic acid-resistant acute promyelocytic leukemia. iScience 2022; 25:103772. [PMID: 35141508 PMCID: PMC8814755 DOI: 10.1016/j.isci.2022.103772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/11/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
All-trans retinoic acid (ATRA) is an essential therapy in the treatment of acute promyelocytic leukemia (APL), but nearly 20% of patients with APL are resistant to ATRA. As there are no biomarkers for ATRA resistance that yet exist, we investigated whether cell mechanics could be associated with this pathological phenotype. Using mechano-node-pore sensing, a single-cell mechanical phenotyping platform, and patient-derived APL cell lines, we discovered that ATRA-resistant APL cells are less mechanically pliable. By investigating how different subcellular components of APL cells contribute to whole-cell mechanical phenotype, we determined that nuclear mechanics strongly influence an APL cell's mechanical response. Moreover, decondensing chromatin with trichostatin A is especially effective in softening ATRA-resistant APL cells. RNA-seq allowed us to compare the transcriptomic differences between ATRA-resistant and ATRA-responsive APL cells and highlighted gene expression changes that could be associated with mechanical changes. Overall, we have demonstrated the potential of "physical" biomarkers in identifying APL resistance.
Collapse
Affiliation(s)
- Brian Li
- UC Berkeley – UCSF Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94709, USA
| | - Annie Maslan
- UC Berkeley – UCSF Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94709, USA
| | - Sean E. Kitayama
- UC Berkeley – UCSF Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94709, USA
| | - Corinne Pierce
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley 94720, USA
| | - Aaron M. Streets
- UC Berkeley – UCSF Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94709, USA
- Center for Computational Biology, University of California, Berkeley, CA 94709, USA
- Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Lydia L. Sohn
- UC Berkeley – UCSF Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94709, USA
- Department of Mechanical Engineering, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
38
|
Zhao Y, Gu L, Sun H, Sha X, Li WJ. Physical Cytometry: Detecting Mass-Related Properties of Single Cells. ACS Sens 2022; 7:21-36. [PMID: 34978200 DOI: 10.1021/acssensors.1c01787] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The physical properties of a single cell, such as mass, volume, and density, are important indications of the cell's metabolic characteristics and homeostasis. Precise measurement of a single cell's mass has long been a challenge due to its minute size. It is only in the past 10 years that a variety of instruments for measuring living cellular mass have emerged with the development of MEMS, microfluidics, and optics technologies. In this review, we discuss the current developments of physical cytometry for quantifying mass-related physical properties of single cells, highlighting the working principle, applications, and unique merits. The review mainly covers these measurement methods: single-cell mass cytometry, levitation image cytometry, suspended microchannel resonator, phase-shifting interferometry, and opto-electrokinetics cell manipulation. Comparisons are made between these methods in terms of throughput, content, invasiveness, compatibility, and precision. Some typical applications of these methods in pathological diagnosis, drug efficacy evaluation, disease treatment, and other related fields are also discussed in this work.
Collapse
Affiliation(s)
- Yuliang Zhao
- School of Control Engineering, Northeastern University, Qinhuangdao 066004, China
| | - Lijia Gu
- School of Control Engineering, Northeastern University, Qinhuangdao 066004, China
| | - Hui Sun
- Department of Mechanical Engineering, City University of Hong Kong, Kowloon, 999077 Hong Kong, China
| | - Xiaopeng Sha
- School of Control Engineering, Northeastern University, Qinhuangdao 066004, China
| | - Wen Jung Li
- Department of Mechanical Engineering, City University of Hong Kong, Kowloon, 999077 Hong Kong, China
| |
Collapse
|
39
|
Cadart C, Venkova L, Piel M, Cosentino Lagomarsino M. Volume growth in animal cells is cell cycle dependent and shows additive fluctuations. eLife 2022; 11:e70816. [PMID: 35088713 PMCID: PMC8798040 DOI: 10.7554/elife.70816] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 12/21/2021] [Indexed: 12/04/2022] Open
Abstract
The way proliferating animal cells coordinate the growth of their mass, volume, and other relevant size parameters is a long-standing question in biology. Studies focusing on cell mass have identified patterns of mass growth as a function of time and cell cycle phase, but little is known about volume growth. To address this question, we improved our fluorescence exclusion method of volume measurement (FXm) and obtained 1700 single-cell volume growth trajectories of HeLa cells. We find that, during most of the cell cycle, volume growth is close to exponential and proceeds at a higher rate in S-G2 than in G1. Comparing the data with a mathematical model, we establish that the cell-to-cell variability in volume growth arises from constant-amplitude fluctuations in volume steps rather than fluctuations of the underlying specific growth rate. We hypothesize that such 'additive noise' could emerge from the processes that regulate volume adaptation to biophysical cues, such as tension or osmotic pressure.
Collapse
Affiliation(s)
- Clotilde Cadart
- Institut Pierre-Gilles de Gennes, PSL Research UniversityParisFrance
- Institut Curie, PSL Research University, CNRSParisFrance
| | - Larisa Venkova
- Institut Pierre-Gilles de Gennes, PSL Research UniversityParisFrance
- Institut Curie, PSL Research University, CNRSParisFrance
| | - Matthieu Piel
- Institut Pierre-Gilles de Gennes, PSL Research UniversityParisFrance
- Institut Curie, PSL Research University, CNRSParisFrance
| | - Marco Cosentino Lagomarsino
- FIRC Institute of Molecular Oncology (IFOM)MilanItaly
- Physics Department, University of Milan, and INFNMilanItaly
| |
Collapse
|
40
|
Kumemura M, Pekin D, Menon VA, Van Seuningen I, Collard D, Tarhan MC. Fabricating Silicon Resonators for Analysing Biological Samples. MICROMACHINES 2021; 12:1546. [PMID: 34945396 PMCID: PMC8708134 DOI: 10.3390/mi12121546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 11/17/2022]
Abstract
The adaptability of microscale devices allows microtechnologies to be used for a wide range of applications. Biology and medicine are among those fields that, in recent decades, have applied microtechnologies to achieve new and improved functionality. However, despite their ability to achieve assay sensitivities that rival or exceed conventional standards, silicon-based microelectromechanical systems remain underutilised for biological and biomedical applications. Although microelectromechanical resonators and actuators do not always exhibit optimal performance in liquid due to electrical double layer formation and high damping, these issues have been solved with some innovative fabrication processes or alternative experimental approaches. This paper focuses on several examples of silicon-based resonating devices with a brief look at their fundamental sensing elements and key fabrication steps, as well as current and potential biological/biomedical applications.
Collapse
Affiliation(s)
- Momoko Kumemura
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu-ku, Kitakyushu-shi, Fukuoka 808-0196, Japan;
- LIMMS/CNRS-IIS, Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; (D.P.); (D.C.)
| | - Deniz Pekin
- LIMMS/CNRS-IIS, Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; (D.P.); (D.C.)
- CNRS/IIS/COL/Lille University, SMMiL-E Project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, CEDEX, 59046 Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France;
| | - Vivek Anand Menon
- Division of Mechanical Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu-shi, Gunma 376-8515, Japan;
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France;
| | - Dominique Collard
- LIMMS/CNRS-IIS, Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; (D.P.); (D.C.)
- CNRS/IIS/COL/Lille University, SMMiL-E Project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, CEDEX, 59046 Lille, France
| | - Mehmet Cagatay Tarhan
- LIMMS/CNRS-IIS, Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan; (D.P.); (D.C.)
- CNRS/IIS/COL/Lille University, SMMiL-E Project, CNRS Délégation Nord-Pas de Calais et Picardie, 2 rue de Canonniers, CEDEX, 59046 Lille, France
- Univ. Lille, CNRS, Centrale Lille, Junia, University Polytechnique Hauts-de-France, UMR 8520—IEMN, Institut
d’Electronique de Microélectronique et de Nanotechnologie, F-59000 Lille, France
| |
Collapse
|
41
|
Tanaka M, Kitanishi-Yumura T, Yumura S. A 'dynamic adder model' for cell size homeostasis in Dictyostelium cells. Sci Rep 2021; 11:13742. [PMID: 34215778 PMCID: PMC8253765 DOI: 10.1038/s41598-021-92700-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
After a cell divides into two daughter cells, the total cell surface area of the daughter cells should increase to the original size to maintain cell size homeostasis in a single cell cycle. Previously, three models have been proposed to explain the regulation of cell size homeostasis: sizer, timer, and adder models. Here, we precisely measured the total cell surface area of Dictyostelium cells in a whole cell cycle by using the agar-overlay method, which eliminated the influence of surface membrane reservoirs, such as microvilli and membrane wrinkles. The total cell surface area exponentially increased during interphase, slightly decreased at metaphase, and then increased by approximately 20% during cytokinesis. From the analysis of the added surface area, we concluded that the cell size was regulated by the adder or near-adder model in interphase. This adder model is not caused by a simple cell membrane addition, but is more dynamic due to the rapid cell membrane turnover. We propose a 'dynamic adder model' to explain cell size homeostasis in interphase.
Collapse
Affiliation(s)
- Masahito Tanaka
- grid.268397.10000 0001 0660 7960Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, 753-8512 Japan ,grid.288127.60000 0004 0466 9350Present Address: Laboratory of Physics and Cell Biology, Department of Chromosome Science, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540 Japan
| | - Toshiko Kitanishi-Yumura
- grid.268397.10000 0001 0660 7960Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, 753-8512 Japan
| | - Shigehiko Yumura
- grid.268397.10000 0001 0660 7960Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, 753-8512 Japan
| |
Collapse
|
42
|
Lee KCM, Guck J, Goda K, Tsia KK. Toward deep biophysical cytometry: prospects and challenges. Trends Biotechnol 2021; 39:1249-1262. [PMID: 33895013 DOI: 10.1016/j.tibtech.2021.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022]
Abstract
The biophysical properties of cells reflect their identities, underpin their homeostatic state in health, and define the pathogenesis of disease. Recent leapfrogging advances in biophysical cytometry now give access to this information, which is obscured in molecular assays, with a discriminative power that was once inconceivable. However, biophysical cytometry should go 'deeper' in terms of exploiting the information-rich cellular biophysical content, generating a molecular knowledge base of cellular biophysical properties, and standardizing the protocols for wider dissemination. Overcoming these barriers, which requires concurrent innovations in microfluidics, optical imaging, and computer vision, could unleash the enormous potential of biophysical cytometry not only for gaining a new mechanistic understanding of biological systems but also for identifying new cost-effective biomarkers of disease.
Collapse
Affiliation(s)
- Kelvin C M Lee
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong
| | - Jochen Guck
- Max Planck Institute for the Science of Light, and Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; Department of Physics, Friedrich-Alexander Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan; Institute of Technological Sciences, Wuhan University, Hubei 430072, China; Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Kevin K Tsia
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong.
| |
Collapse
|
43
|
Liu X, Oh S, Peshkin L, Kirschner MW. Computationally enhanced quantitative phase microscopy reveals autonomous oscillations in mammalian cell growth. Proc Natl Acad Sci U S A 2020; 117:27388-27399. [PMID: 33087574 PMCID: PMC7959529 DOI: 10.1073/pnas.2002152117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The fine balance of growth and division is a fundamental property of the physiology of cells, and one of the least understood. Its study has been thwarted by difficulties in the accurate measurement of cell size and the even greater challenges of measuring growth of a single cell over time. We address these limitations by demonstrating a computationally enhanced methodology for quantitative phase microscopy for adherent cells, using improved image processing algorithms and automated cell-tracking software. Accuracy has been improved more than twofold and this improvement is sufficient to establish the dynamics of cell growth and adherence to simple growth laws. It is also sufficient to reveal unknown features of cell growth, previously unmeasurable. With these methodological and analytical improvements, in several cell lines we document a remarkable oscillation in growth rate, occurring throughout the cell cycle, coupled to cell division or birth yet independent of cell cycle progression. We expect that further exploration with this advanced tool will provide a better understanding of growth rate regulation in mammalian cells.
Collapse
Affiliation(s)
- Xili Liu
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Seungeun Oh
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Leonid Peshkin
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
44
|
Monitoring and modeling of lymphocytic leukemia cell bioenergetics reveals decreased ATP synthesis during cell division. Nat Commun 2020; 11:4983. [PMID: 33020492 PMCID: PMC7536222 DOI: 10.1038/s41467-020-18769-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
The energetic demands of a cell are believed to increase during mitosis, but the rates of ATP synthesis and consumption during mitosis have not been quantified. Here, we monitor mitochondrial membrane potential of single lymphocytic leukemia cells and demonstrate that mitochondria hyperpolarize from the G2/M transition until the metaphase-anaphase transition. This hyperpolarization was dependent on cyclin-dependent kinase 1 (CDK1) activity. By using an electrical circuit model of mitochondria, we quantify mitochondrial ATP synthesis rates in mitosis from the single-cell time-dynamics of mitochondrial membrane potential. We find that mitochondrial ATP synthesis decreases by approximately 50% during early mitosis and increases back to G2 levels during cytokinesis. Consistently, ATP levels and ATP synthesis are lower in mitosis than in G2 in synchronized cell populations. Overall, our results provide insights into mitotic bioenergetics and suggest that cell division is not a highly energy demanding process. ATP drives most cellular processes, although ATP production and consumption levels during mitosis remain unreported. Here, the authors combine metabolic measurements and modeling to quantify ATP levels and synthesis dynamics, revealing that ATP synthesis and consumption are lowered during mitosis.
Collapse
|