1
|
Dixon JC, Frick CL, Leveille CL, Garrison P, Lee PA, Mogre SS, Morris B, Nivedita N, Vasan R, Chen J, Fraser CL, Gamlin CR, Harris LK, Hendershott MC, Johnson GT, Klein KN, Oluoch SA, Thirstrup DJ, Sluzewski MF, Wilhelm L, Yang R, Toloudis DM, Viana MP, Theriot JA, Rafelski SM. Colony context and size-dependent compensation mechanisms give rise to variations in nuclear growth trajectories. Cell Syst 2025; 16:101265. [PMID: 40315848 DOI: 10.1016/j.cels.2025.101265] [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] [Received: 07/24/2024] [Revised: 12/10/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
To investigate how cellular variations arise across spatiotemporal scales in a population of identical healthy cells, we performed a data-driven analysis of nuclear growth variations in hiPS cell colonies as a model system. We generated a 3D timelapse dataset of thousands of nuclei over multiple days and developed open-source tools for image and data analysis and feature-based timelapse data exploration. Together, these data, tools, and workflows comprise a framework for systematic quantitative analysis of dynamics at individual and population levels, and the analysis further highlights important aspects to consider when interpreting timelapse data. We found that individual nuclear volume growth trajectories arise from short-timescale variations attributable to their spatiotemporal context within the colony. We identified a time-invariant volume compensation relationship between nuclear growth duration and starting volume across the population. Notably, we discovered that inheritance plays a crucial role in determining these two key nuclear growth features while other growth features are determined by their spatiotemporal context and are not inherited.
Collapse
Affiliation(s)
- Julie C Dixon
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Christopher L Frick
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | | | - Philip Garrison
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Peyton A Lee
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Saurabh S Mogre
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Benjamin Morris
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Nivedita Nivedita
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Ritvik Vasan
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Jianxu Chen
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Cameron L Fraser
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Clare R Gamlin
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Leigh K Harris
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | | | - Graham T Johnson
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Kyle N Klein
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Sandra A Oluoch
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Derek J Thirstrup
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - M Filip Sluzewski
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Lyndsay Wilhelm
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Ruian Yang
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Daniel M Toloudis
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Matheus P Viana
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Susanne M Rafelski
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA 98109, USA.
| |
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
|
Cano Á, Yubero ML, Millá C, Puerto-Belda V, Ruz JJ, Kosaka PM, Calleja M, Malumbres M, Tamayo J. Rapid mechanical phenotyping of breast cancer cells based on stochastic intracellular fluctuations. iScience 2024; 27:110960. [PMID: 39493877 PMCID: PMC11530848 DOI: 10.1016/j.isci.2024.110960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/16/2024] [Accepted: 09/11/2024] [Indexed: 11/05/2024] Open
Abstract
Predicting the phenotypic impact of genetic variants and treatments is crucial in cancer genetics and precision oncology. Here, we have developed a noise decorrelation method that enables quantitative phase imaging (QPI) with the capability for label-free noninvasive mapping of intracellular dry mass fluctuations within the millisecond-to-second timescale regime, previously inaccessible due to temporal phase noise. Applied to breast cancer cells, this method revealed regions driven by thermal forces and regions of intense activity fueled by ATP hydrolysis. Intriguingly, as malignancy increases, the cells strategically expand these active regions to satisfy increasing energy demands. We propose parameters encapsulating key information about the spatiotemporal distribution of intracellular fluctuations, enabling precise phenotyping. This technique addresses the need for accurate, rapid functional screening methods in cancer medicine.
Collapse
Affiliation(s)
- Álvaro Cano
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| | - Marina L. Yubero
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| | - Carmen Millá
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| | - Verónica Puerto-Belda
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| | - Jose J. Ruz
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| | - Priscila M. Kosaka
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| | - Montserrat Calleja
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| | - Marcos Malumbres
- Cancer Cell Cycle Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Javier Tamayo
- Bionanomechanics Lab, Instituto de Micro y Nanotecnología, IMN-CNM (CSIC), Tres Cantos, Madrid, Spain
| |
Collapse
|
4
|
Zhang J, Griffin J, Roy K, Hoffmann A, Zangle TA. Tracking of lineage mass via quantitative phase imaging and confinement in low refractive index microwells. LAB ON A CHIP 2024; 24:4440-4449. [PMID: 39190401 PMCID: PMC11412070 DOI: 10.1039/d4lc00389f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Measurements of cell lineages are central to a variety of fundamental biological questions, ranging from developmental to cancer biology. However, accurate lineage tracing requires nearly perfect cell tracking, which can be challenging due to cell motion during imaging. Here we demonstrate the integration of microfabrication, imaging, and image processing approaches to demonstrate a platform for cell lineage tracing. We use quantitative phase imaging (QPI), a label-free imaging approach that quantifies cell mass. This gives an additional parameter, cell mass, that can be used to improve tracking accuracy. We confine lineages within microwells fabricated to reduce cell adhesion to sidewalls made of a low refractive index polymer. This also allows the microwells themselves to serve as references for QPI, enabling measurement of cell mass even in confluent microwells. We demonstrate application of this approach to immortalized adherent and nonadherent cell lines as well as stimulated primary B cells cultured ex vivo. Overall, our approach enables lineage tracking, or measurement of lineage mass, in a platform that can be customized to varied cell types.
Collapse
Affiliation(s)
- Jingzhou Zhang
- Department of Chemical Engineering, University of Utah, USA.
| | - Justin Griffin
- Department of Chemical Engineering, University of Utah, USA.
| | - Koushik Roy
- Division of Microbiology and Immunology, Department of Pathology, School of Medicine, University of Utah, USA
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Institute for Quantitative and Computational Biosciences, and Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Thomas A Zangle
- Department of Chemical Engineering, University of Utah, USA.
- Huntsman Cancer Institute, University of Utah, USA
| |
Collapse
|
5
|
Iida S, Ide S, Tamura S, Sasai M, Tani T, Goto T, Shribak M, Maeshima K. Orientation-independent-DIC imaging reveals that a transient rise in depletion attraction contributes to mitotic chromosome condensation. Proc Natl Acad Sci U S A 2024; 121:e2403153121. [PMID: 39190347 PMCID: PMC11388287 DOI: 10.1073/pnas.2403153121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/19/2024] [Indexed: 08/28/2024] Open
Abstract
Genomic information must be faithfully transmitted into two daughter cells during mitosis. To ensure the transmission process, interphase chromatin is further condensed into mitotic chromosomes. Although protein factors like condensins and topoisomerase IIα are involved in the assembly of mitotic chromosomes, the physical bases of the condensation process remain unclear. Depletion attraction/macromolecular crowding, an effective attractive force that arises between large structures in crowded environments around chromosomes, may contribute to the condensation process. To approach this issue, we investigated the "chromosome milieu" during mitosis of living human cells using an orientation-independent-differential interference contrast module combined with a confocal laser scanning microscope, which is capable of precisely mapping optical path differences and estimating molecular densities. We found that the molecular density surrounding chromosomes increased with the progression from prophase to anaphase, concurring with chromosome condensation. However, the molecular density went down in telophase, when chromosome decondensation began. Changes in the molecular density around chromosomes by hypotonic or hypertonic treatment consistently altered the condensation levels of chromosomes. In vitro, native chromatin was converted into liquid droplets of chromatin in the presence of cations and a macromolecular crowder. Additional crowder made the chromatin droplets stiffer and more solid-like. These results suggest that a transient rise in depletion attraction, likely triggered by the relocation of macromolecules (proteins, RNAs, and others) via nuclear envelope breakdown and by a subsequent decrease in cell volumes, contributes to mitotic chromosome condensation, shedding light on a different aspect of the condensation mechanism in living human cells.
Collapse
Affiliation(s)
- Shiori Iida
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka411-8540, Japan
- Graduate Institute for Advanced Studies (SOKENDAI), Mishima, Shizuoka411-8540, Japan
| | - Satoru Ide
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka411-8540, Japan
- Graduate Institute for Advanced Studies (SOKENDAI), Mishima, Shizuoka411-8540, Japan
| | - Sachiko Tamura
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka411-8540, Japan
| | - Masaki Sasai
- Fukui Institute for Fundamental Chemistry, Kyoto University, Kyoto606-8103, Japan
- Department of Complex Systems Science, Nagoya University, Nagoya464-8603, Japan
| | - Tomomi Tani
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ikeda, Osaka563-8577, Japan
| | - Tatsuhiko Goto
- Research Center for Global Agromedicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido080-8555, Japan
- Department of Life and Food Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido080-8555, Japan
| | | | - Kazuhiro Maeshima
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka411-8540, Japan
- Graduate Institute for Advanced Studies (SOKENDAI), Mishima, Shizuoka411-8540, Japan
| |
Collapse
|
6
|
Dixon JC, Frick CL, Leveille CL, Garrison P, Lee PA, Mogre SS, Morris B, Nivedita N, Vasan R, Chen J, Fraser CL, Gamlin CR, Harris LK, Hendershott MC, Johnson GT, Klein KN, Oluoch SA, Thirstrup DJ, Sluzewski MF, Wilhelm L, Yang R, Toloudis DM, Viana MP, Theriot JA, Rafelski SM. Colony context and size-dependent compensation mechanisms give rise to variations in nuclear growth trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601071. [PMID: 38979140 PMCID: PMC11230432 DOI: 10.1101/2024.06.28.601071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
To investigate the fundamental question of how cellular variations arise across spatiotemporal scales in a population of identical healthy cells, we focused on nuclear growth in hiPS cell colonies as a model system. We generated a 3D timelapse dataset of thousands of nuclei over multiple days, and developed open-source tools for image and data analysis and an interactive timelapse viewer for exploring quantitative features of nuclear size and shape. We performed a data-driven analysis of nuclear growth variations across timescales. We found that individual nuclear volume growth trajectories arise from short timescale variations attributable to their spatiotemporal context within the colony. We identified a strikingly time-invariant volume compensation relationship between nuclear growth duration and starting volume across the population. Notably, we discovered that inheritance plays a crucial role in determining these two key nuclear growth features while other growth features are determined by their spatiotemporal context and are not inherited.
Collapse
Affiliation(s)
- Julie C. Dixon
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Christopher L. Frick
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Chantelle L. Leveille
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Philip Garrison
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Peyton A. Lee
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Saurabh S. Mogre
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Benjamin Morris
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Nivedita Nivedita
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Ritvik Vasan
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- These authors contributed equally to this work
| | - Jianxu Chen
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
- Present address: Leibniz-Institut fur Analytische Wissenschaften – ISAS – e.V., Dortmund, 44139, Germany
| | - Cameron L. Fraser
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Clare R. Gamlin
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Leigh K. Harris
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | | | - Graham T. Johnson
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Kyle N. Klein
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Sandra A. Oluoch
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Derek J. Thirstrup
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - M. Filip Sluzewski
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Lyndsay Wilhelm
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Ruian Yang
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Daniel M. Toloudis
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Matheus P. Viana
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| | - Julie A. Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Susanne M. Rafelski
- Allen Institute for Cell Science, 615 Westlake Ave N, Seattle, WA, 98109, USA
| |
Collapse
|
7
|
Liu Y, Uttam S. Perspective on quantitative phase imaging to improve precision cancer medicine. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:S22705. [PMID: 38584967 PMCID: PMC10996848 DOI: 10.1117/1.jbo.29.s2.s22705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/03/2024] [Accepted: 03/15/2024] [Indexed: 04/09/2024]
Abstract
Significance Quantitative phase imaging (QPI) offers a label-free approach to non-invasively characterize cellular processes by exploiting their refractive index based intrinsic contrast. QPI captures this contrast by translating refractive index associated phase shifts into intensity-based quantifiable data with nanoscale sensitivity. It holds significant potential for advancing precision cancer medicine by providing quantitative characterization of the biophysical properties of cells and tissue in their natural states. Aim This perspective aims to discuss the potential of QPI to increase our understanding of cancer development and its response to therapeutics. It also explores new developments in QPI methods towards advancing personalized cancer therapy and early detection. Approach We begin by detailing the technical advancements of QPI, examining its implementations across transmission and reflection geometries and phase retrieval methods, both interferometric and non-interferometric. The focus then shifts to QPI's applications in cancer research, including dynamic cell mass imaging for drug response assessment, cancer risk stratification, and in-vivo tissue imaging. Results QPI has emerged as a crucial tool in precision cancer medicine, offering insights into tumor biology and treatment efficacy. Its sensitivity to detecting nanoscale changes holds promise for enhancing cancer diagnostics, risk assessment, and prognostication. The future of QPI is envisioned in its integration with artificial intelligence, morpho-dynamics, and spatial biology, broadening its impact in cancer research. Conclusions QPI presents significant potential in advancing precision cancer medicine and redefining our approach to cancer diagnosis, monitoring, and treatment. Future directions include harnessing high-throughput dynamic imaging, 3D QPI for realistic tumor models, and combining artificial intelligence with multi-omics data to extend QPI's capabilities. As a result, QPI stands at the forefront of cancer research and clinical application in cancer care.
Collapse
Affiliation(s)
- Yang Liu
- University of Illinois Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Cancer Center at Illinois, Department of Bioengineering, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
- University of Pittsburgh, Departments of Medicine and Bioengineering, Pittsburgh, Pennsylvania, United States
| | - Shikhar Uttam
- University of Pittsburgh, Department of Computational and Systems Biology, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
8
|
Hur SW, Kwon M, Manoharaan R, Mohammadi MH, Samuel AZ, Mulligan MP, Hergenrother PJ, Bhargava R. Capturing cell morphology dynamics with high temporal resolution using single-shot quantitative phase gradient imaging. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:S22712. [PMID: 39015510 PMCID: PMC11249975 DOI: 10.1117/1.jbo.29.s2.s22712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/18/2024]
Abstract
Significance Label-free quantitative phase imaging can potentially measure cellular dynamics with minimal perturbation, motivating efforts to develop faster and more sensitive instrumentation. We characterize fast, single-shot quantitative phase gradient microscopy (ss-QPGM) that simultaneously acquires multiple polarization components required to reconstruct phase images. We integrate a computationally efficient least squares algorithm to provide real-time, video-rate imaging (up to 75 frames / s ). The developed instrument was used to observe changes in cellular morphology and correlate these to molecular measures commonly obtained by staining. Aim We aim to characterize a fast approach to ss-QPGM and record morphological changes in single-cell phase images. We also correlate these with biochemical changes indicating cell death using concurrently acquired fluorescence images. Approach Here, we examine nutrient deprivation and anticancer drug-induced cell death in two different breast cell lines, viz., M2 and MCF7. Our approach involves in-line measurements of ss-QPGM and fluorescence imaging of the cells biochemically labeled for viability. Results We validate the accuracy of the phase measurement using a USAF1951 pattern phase target. The ss-QPGM system resolves 912.3 lp / mm , and our analysis scheme accurately retrieves the phase with a high correlation coefficient ( ∼ 0.99 ), as measured by calibrated sample thicknesses. Analyzing the contrast in phase, we estimate the spatial resolution achievable to be 0.55 μ m for this microscope. ss-QPGM time-lapse live-cell imaging reveals multiple intracellular and morphological changes during biochemically induced cell death. Inferences from co-registered images of quantitative phase and fluorescence suggest the possibility of necrosis, which agrees with previous findings. Conclusions Label-free ss-QPGM with high-temporal resolution and high spatial fidelity is demonstrated. Its application for monitoring dynamic changes in live cells offers promising prospects.
Collapse
Affiliation(s)
- Sun Woong Hur
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Urbana, Illinois, United States
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
| | - Minsung Kwon
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Urbana, Illinois, United States
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
| | - Revathi Manoharaan
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Urbana, Illinois, United States
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
| | - Melika Haji Mohammadi
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Urbana, Illinois, United States
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
| | - Ashok Zachariah Samuel
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Urbana, Illinois, United States
| | - Michael P. Mulligan
- University of Illinois at Urbana-Champaign, Department of Chemistry, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Carl R. Woese Institute for Genomic Biology, Urbana, Illinois, United States
| | - Paul J. Hergenrother
- University of Illinois at Urbana-Champaign, Department of Chemistry, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Carl R. Woese Institute for Genomic Biology, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Cancer Center at Illinois, Urbana, Illinois, United States
| | - Rohit Bhargava
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Urbana, Illinois, United States
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Carl R. Woese Institute for Genomic Biology, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Cancer Center at Illinois, Urbana, Illinois, United States
- University of Illinois Urbana-Champaign, Department of Chemical and Biomolecular Engineering, Electrical and Computer Engineering, Mechanical Science and Engineering and Chemistry, Urbana, Illinois, United States
| |
Collapse
|
9
|
Pradeep S, Zangle TA. LVING reveals the intracellular structure of cell growth. Sci Rep 2024; 14:8544. [PMID: 38609444 PMCID: PMC11014851 DOI: 10.1038/s41598-024-58992-x] [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: 11/12/2023] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
The continuous balance of growth and degradation inside cells maintains homeostasis. Disturbance of this balance by internal or external factors cause state of disease, while effective disease treatments seek to restore this balance. Here, we present a method based on quantitative phase imaging (QPI) based measurements of cell mass and the velocity of mass transport to quantify the balance of growth and degradation within intracellular control volumes. The result, which we call Lagrangian velocimetry for intracellular net growth (LVING), provides high resolution maps of intracellular biomass production and degradation. We use LVING to quantify the growth in different regions of the cell during phases of the cell cycle. LVING can also be used to quantitatively compare the effect of range of chemotherapy drug doses on subcellular growth processes. Finally, we applied LVING to characterize the effect of autophagy on the growth machinery inside cells. Overall, LVING reveals both the structure and distribution of basal growth within cells, as well as the disruptions to this structure that occur during alterations in cell state.
Collapse
Affiliation(s)
- Soorya Pradeep
- Department of Chemical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Thomas A Zangle
- Department of Chemical Engineering, University of Utah, Salt Lake City, Utah, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
10
|
Chen Y, Huang JH, Phong C, Ferrell JE. Viscosity-dependent control of protein synthesis and degradation. Nat Commun 2024; 15:2149. [PMID: 38459041 PMCID: PMC10923802 DOI: 10.1038/s41467-024-46447-w] [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: 10/29/2023] [Accepted: 02/28/2024] [Indexed: 03/10/2024] Open
Abstract
It has been proposed that the concentration of proteins in the cytoplasm maximizes the speed of important biochemical reactions. Here we have used Xenopus egg extracts, which can be diluted or concentrated to yield a range of cytoplasmic protein concentrations, to test the effect of cytoplasmic concentration on mRNA translation and protein degradation. We find that protein synthesis rates are maximal in ~1x cytoplasm, whereas protein degradation continues to rise to a higher optimal concentration of ~1.8x. We show that this difference in optima can be attributed to a greater sensitivity of translation to cytoplasmic viscosity. The different concentration optima could produce a negative feedback homeostatic system, where increasing the cytoplasmic protein concentration above the 1x physiological level increases the viscosity of the cytoplasm, which selectively inhibits translation and drives the system back toward the 1x set point.
Collapse
Affiliation(s)
- Yuping Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Jo-Hsi Huang
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Connie Phong
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - James E Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
11
|
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: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
12
|
Liu X, Moscvin M, Oh S, Chen T, Choi W, Evans B, Rowell SM, Nadeem O, Mo CC, Sperling AS, Anderson KC, Yaqoob Z, Bianchi G, Sung Y. Characterizing dry mass and volume changes in human multiple myeloma cells upon treatment with proteotoxic and genotoxic drugs. Clin Exp Med 2023; 23:3821-3832. [PMID: 37421589 PMCID: PMC10777533 DOI: 10.1007/s10238-023-01124-y] [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: 03/23/2023] [Accepted: 06/20/2023] [Indexed: 07/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of terminally differentiated plasma cells. MM remains incurable, but overall survival of patients has progressively increased over the past two decades largely due to novel agents such as proteasome inhibitors (PI) and the immunomodulatory agents. While these therapies are highly effective, MM patients can be de novo resistant and acquired resistance with prolonged treatment is inevitable. There is growing interest in early, accurate identification of responsive versus non-responsive patients; however, limited sample availability and need for rapid assays are limiting factors. Here, we test dry mass and volume as label-free biomarkers to monitor early response of MM cells to treatment with bortezomib, doxorubicin, and ultraviolet light. For the dry mass measurement, we use two types of phase-sensitive optical microscopy techniques: digital holographic tomography and computationally enhanced quantitative phase microscopy. We show that human MM cell lines (RPMI8226, MM.1S, KMS20, and AMO1) increase dry mass upon bortezomib treatment. This dry mass increase after bortezomib treatment occurs as early as 1 h for sensitive cells and 4 h for all tested cells. We further confirm this observation using primary multiple myeloma cells derived from patients and show that a correlation exists between increase in dry mass and sensitivity to bortezomib, supporting the use of dry mass as a biomarker. The volume measurement using Coulter counter shows a more complex behavior; RPMI8226 cells increase the volume at an early stage of apoptosis, but MM.1S cells show the volume decrease typically observed with apoptotic cells. Altogether, this cell study presents complex kinetics of dry mass and volume at an early stage of apoptosis, which may serve as a basis for the detection and treatment of MM cells.
Collapse
Affiliation(s)
- Xili Liu
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Maria Moscvin
- Amyloidosis Program, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Seungeun Oh
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Tianzeng Chen
- Amyloidosis Program, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wonshik Choi
- Department of Physics, Korea University, Seoul, Korea
| | - Benjamin Evans
- Amyloidosis Program, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sean M Rowell
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Omar Nadeem
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Clifton C Mo
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Adam S Sperling
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kenneth C Anderson
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, 02115, USA
| | - Zahid Yaqoob
- Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Giada Bianchi
- Amyloidosis Program, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Yongjin Sung
- College of Engineering and Applied Science, University of Wisconsin, Milwaukee, WI, 53211, USA.
| |
Collapse
|
13
|
Tanwar S, Wu L, Zahn N, Raj P, Ghaemi B, Chatterjee A, Bulte JWM, Barman I. Targeted Enzyme Activity Imaging with Quantitative Phase Microscopy. NANO LETTERS 2023; 23:4602-4608. [PMID: 37154678 PMCID: PMC10798004 DOI: 10.1021/acs.nanolett.3c01090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Quantitative phase imaging (QPI) is a powerful optical imaging modality for label-free, rapid, and three-dimensional (3D) monitoring of cells and tissues. However, molecular imaging of important intracellular biomolecules such as enzymes remains a largely unexplored area for QPI. Herein, we introduce a fundamentally new approach by designing QPI contrast agents that allow sensitive detection of intracellular biomolecules. We report a new class of bio-orthogonal QPI-nanoprobes for in situ high-contrast refractive index (RI) imaging of enzyme activity. The nanoprobes feature silica nanoparticles (SiO2 NPs) having higher RI than endogenous cellular components and surface-anchored cyanobenzothiazole-cysteine (CBT-Cys) conjugated enzyme-responsive peptide sequences. The nanoprobes specifically aggregated in cells with target enzyme activity, increasing intracellular RI and enabling precise visualization of intracellular enzyme activity. We envision that this general design of QPI-nanoprobes could open doors for spatial-temporal mapping of enzyme activity with direct implications for disease diagnosis and evaluating the therapeutic efficacy.
Collapse
Affiliation(s)
- Swati Tanwar
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Noah Zahn
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Behnaz Ghaemi
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Arnab Chatterjee
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Jeff W M Bulte
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Inc., Baltimore, Maryland 21205, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| |
Collapse
|
14
|
Chen Y, Huang JH, Phong C, Ferrell JE. Protein homeostasis from diffusion-dependent control of protein synthesis and degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.538146. [PMID: 37162886 PMCID: PMC10168264 DOI: 10.1101/2023.04.24.538146] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
It has been proposed that the concentration of proteins in the cytoplasm maximizes the speed of important biochemical reactions. Here we have used the Xenopus extract system, which can be diluted or concentrated to yield a range of cytoplasmic protein concentrations, to test the effect of cytoplasmic concentration on mRNA translation and protein degradation. We found that protein synthesis rates are maximal in ~1x cytoplasm, whereas protein degradation continues to rise to an optimal concentration of ~1.8x. This can be attributed to the greater sensitivity of translation to cytoplasmic viscosity, perhaps because it involves unusually large macromolecular complexes like polyribosomes. The different concentration optima sets up a negative feedback homeostatic system, where increasing the cytoplasmic protein concentration above the 1x physiological level increases the viscosity of the cytoplasm, which selectively inhibits translation and drives the system back toward the 1x set point.
Collapse
Affiliation(s)
- Yuping Chen
- Dept. of Chemical and Systems Biology, Stanford University School of Medicine, Stanford CA 94305
- These authors contributed equally
- Corresponding authors
| | - Jo-Hsi Huang
- Dept. of Chemical and Systems Biology, Stanford University School of Medicine, Stanford CA 94305
- These authors contributed equally
| | - Connie Phong
- Dept. of Chemical and Systems Biology, Stanford University School of Medicine, Stanford CA 94305
| | - James E. Ferrell
- Dept. of Chemical and Systems Biology, Stanford University School of Medicine, Stanford CA 94305
- Dept. of Biochemistry, Stanford University School of Medicine, Stanford CA 94305
- Corresponding authors
- Lead contact
| |
Collapse
|
15
|
Takhaveev V, Özsezen S, Smith EN, Zylstra A, Chaillet ML, Chen H, Papagiannakis A, Milias-Argeitis A, Heinemann M. Temporal segregation of biosynthetic processes is responsible for metabolic oscillations during the budding yeast cell cycle. Nat Metab 2023; 5:294-313. [PMID: 36849832 PMCID: PMC9970877 DOI: 10.1038/s42255-023-00741-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 01/10/2023] [Indexed: 03/01/2023]
Abstract
Many cell biological and biochemical mechanisms controlling the fundamental process of eukaryotic cell division have been identified; however, the temporal dynamics of biosynthetic processes during the cell division cycle are still elusive. Here, we show that key biosynthetic processes are temporally segregated along the cell cycle. Using budding yeast as a model and single-cell methods to dynamically measure metabolic activity, we observe two peaks in protein synthesis, in the G1 and S/G2/M phase, whereas lipid and polysaccharide synthesis peaks only once, during the S/G2/M phase. Integrating the inferred biosynthetic rates into a thermodynamic-stoichiometric metabolic model, we find that this temporal segregation in biosynthetic processes causes flux changes in primary metabolism, with an acceleration of glucose-uptake flux in G1 and phase-shifted oscillations of oxygen and carbon dioxide exchanges. Through experimental validation of the model predictions, we demonstrate that primary metabolism oscillates with cell-cycle periodicity to satisfy the changing demands of biosynthetic processes exhibiting unexpected dynamics during the cell cycle.
Collapse
Affiliation(s)
- Vakil Takhaveev
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Serdar Özsezen
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Edward N Smith
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Andre Zylstra
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Marten L Chaillet
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Haoqi Chen
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Alexandros Papagiannakis
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
- Department of Biology and Sarafan Chemistry, Engineering, and Medicine for Human Health Institute, Stanford University, Stanford, CA, USA
| | - Andreas Milias-Argeitis
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Matthias Heinemann
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
16
|
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: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [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
|
17
|
Liu X, Oh S, Kirschner MW. The uniformity and stability of cellular mass density in mammalian cell culture. Front Cell Dev Biol 2022; 10:1017499. [PMID: 36313562 PMCID: PMC9597509 DOI: 10.3389/fcell.2022.1017499] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
Cell dry mass is principally determined by the sum of biosynthesis and degradation. Measurable change in dry mass occurs on a time scale of hours. By contrast, cell volume can change in minutes by altering the osmotic conditions. How changes in dry mass and volume are coupled is a fundamental question in cell size control. If cell volume were proportional to cell dry mass during growth, the cell would always maintain the same cellular mass density, defined as cell dry mass dividing by cell volume. The accuracy and stability against perturbation of this proportionality has never been stringently tested. Normalized Raman Imaging (NoRI), can measure both protein and lipid dry mass density directly. Using this new technique, we have been able to investigate the stability of mass density in response to pharmaceutical and physiological perturbations in three cultured mammalian cell lines. We find a remarkably narrow mass density distribution within cells, that is, significantly tighter than the variability of mass or volume distribution. The measured mass density is independent of the cell cycle. We find that mass density can be modulated directly by extracellular osmolytes or by disruptions of the cytoskeleton. Yet, mass density is surprisingly resistant to pharmacological perturbations of protein synthesis or protein degradation, suggesting there must be some form of feedback control to maintain the homeostasis of mass density when mass is altered. By contrast, physiological perturbations such as starvation or senescence induce significant shifts in mass density. We have begun to shed light on how and why cell mass density remains fixed against some perturbations and yet is sensitive during transitions in physiological state.
Collapse
Affiliation(s)
| | | | - Marc W. Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
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
|
19
|
Concentration fluctuations in growing and dividing cells: Insights into the emergence of concentration homeostasis. PLoS Comput Biol 2022; 18:e1010574. [PMID: 36194626 PMCID: PMC9565450 DOI: 10.1371/journal.pcbi.1010574] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/14/2022] [Accepted: 09/14/2022] [Indexed: 11/19/2022] Open
Abstract
Intracellular reaction rates depend on concentrations and hence their levels are often regulated. However classical models of stochastic gene expression lack a cell size description and cannot be used to predict noise in concentrations. Here, we construct a model of gene product dynamics that includes a description of cell growth, cell division, size-dependent gene expression, gene dosage compensation, and size control mechanisms that can vary with the cell cycle phase. We obtain expressions for the approximate distributions and power spectra of concentration fluctuations which lead to insight into the emergence of concentration homeostasis. We find that (i) the conditions necessary to suppress cell division-induced concentration oscillations are difficult to achieve; (ii) mRNA concentration and number distributions can have different number of modes; (iii) two-layer size control strategies such as sizer-timer or adder-timer are ideal because they maintain constant mean concentrations whilst minimising concentration noise; (iv) accurate concentration homeostasis requires a fine tuning of dosage compensation, replication timing, and size-dependent gene expression; (v) deviations from perfect concentration homeostasis show up as deviations of the concentration distribution from a gamma distribution. Some of these predictions are confirmed using data for E. coli, fission yeast, and budding yeast.
Collapse
|
20
|
Nguyen TL, Pradeep S, Judson-Torres RL, Reed J, Teitell MA, Zangle TA. Quantitative Phase Imaging: Recent Advances and Expanding Potential in Biomedicine. ACS NANO 2022; 16:11516-11544. [PMID: 35916417 PMCID: PMC10112851 DOI: 10.1021/acsnano.1c11507] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Quantitative phase imaging (QPI) is a label-free, wide-field microscopy approach with significant opportunities for biomedical applications. QPI uses the natural phase shift of light as it passes through a transparent object, such as a mammalian cell, to quantify biomass distribution and spatial and temporal changes in biomass. Reported in cell studies more than 60 years ago, ongoing advances in QPI hardware and software are leading to numerous applications in biology, with a dramatic expansion in utility over the past two decades. Today, investigations of cell size, morphology, behavior, cellular viscoelasticity, drug efficacy, biomass accumulation and turnover, and transport mechanics are supporting studies of development, physiology, neural activity, cancer, and additional physiological processes and diseases. Here, we review the field of QPI in biology starting with underlying principles, followed by a discussion of technical approaches currently available or being developed, and end with an examination of the breadth of applications in use or under development. We comment on strengths and shortcomings for the deployment of QPI in key biomedical contexts and conclude with emerging challenges and opportunities based on combining QPI with other methodologies that expand the scope and utility of QPI even further.
Collapse
|
21
|
Guerra P, Vuillemenot LAPE, van Oppen YB, Been M, Milias-Argeitis A. TORC1 and PKA activity towards ribosome biogenesis oscillates in synchrony with the budding yeast cell cycle. J Cell Sci 2022; 135:276358. [PMID: 35975715 DOI: 10.1242/jcs.260378] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/11/2022] [Indexed: 10/15/2022] Open
Abstract
Recent studies have revealed that the growth rate of budding yeast and mammalian cells varies during the cell cycle. By linking a multitude of signals to cell growth, the highly conserved Target of Rapamycin Complex 1 (TORC1) and Protein Kinase A (PKA) pathways are prime candidates for mediating the dynamic coupling between growth and division. However, measurements of TORC1 and PKA activity during the cell cycle are still lacking. Following the localization dynamics of two TORC1 and PKA targets via time-lapse microscopy in hundreds of yeast cells, we found that the activity of these pathways towards ribosome biogenesis fluctuates in synchrony with the cell cycle even under constant external conditions. Mutations of upstream TORC1 and PKA regulators suggested that internal metabolic signals partially mediate these activity changes. Our study reveals a new aspect of TORC1 and PKA signaling, which will be important for understanding growth regulation during the cell cycle.
Collapse
Affiliation(s)
- Paolo Guerra
- Molecular Systems Biology, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Netherlands
| | - Luc-Alban P E Vuillemenot
- Molecular Systems Biology, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Netherlands
| | - Yulan B van Oppen
- Molecular Systems Biology, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Netherlands
| | - Marije Been
- Molecular Systems Biology, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Netherlands
| | - Andreas Milias-Argeitis
- Molecular Systems Biology, Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Netherlands
| |
Collapse
|
22
|
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: 20] [Impact Index Per Article: 6.7] [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
|
23
|
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
|
24
|
Wu Y, Pegoraro AF, Weitz DA, Janmey P, Sun SX. The correlation between cell and nucleus size is explained by an eukaryotic cell growth model. PLoS Comput Biol 2022; 18:e1009400. [PMID: 35180215 PMCID: PMC8893647 DOI: 10.1371/journal.pcbi.1009400] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 03/03/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
In eukaryotes, the cell volume is observed to be strongly correlated with the nuclear volume. The slope of this correlation depends on the cell type, growth condition, and the physical environment of the cell. We develop a computational model of cell growth and proteome increase, incorporating the kinetics of amino acid import, protein/ribosome synthesis and degradation, and active transport of proteins between the cytoplasm and the nucleoplasm. We also include a simple model of ribosome biogenesis and assembly. Results show that the cell volume is tightly correlated with the nuclear volume, and the cytoplasm-nucleoplasm transport rates strongly influence the cell growth rate as well as the cell/nucleus volume ratio (C/N ratio). Ribosome assembly and the ratio of ribosomal proteins to mature ribosomes also influence the cell volume and the cell growth rate. We find that in order to regulate the cell growth rate and the cell/nucleus volume ratio, the cell must optimally control groups of kinetic and transport parameters together, which could explain the quantitative roles of canonical growth pathways. Finally, although not explicitly demonstrated in this work, we point out that it is possible to construct a detailed proteome distribution using our model and RNAseq data, provided that a quantitative cell division mechanism is known.
Collapse
Affiliation(s)
- Yufei Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | | | - David A. Weitz
- Department of Physics, Harvard University, Boston, Massachusetts, United States of America
| | - Paul Janmey
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Sean X. Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Mofatteh M, Echegaray-Iturra F, Alamban A, Dalla Ricca F, Bakshi A, Aydogan MG. Autonomous clocks that regulate organelle biogenesis, cytoskeletal organization, and intracellular dynamics. eLife 2021; 10:e72104. [PMID: 34586070 PMCID: PMC8480978 DOI: 10.7554/elife.72104] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/14/2021] [Indexed: 12/27/2022] Open
Abstract
How do cells perceive time? Do cells use temporal information to regulate the production/degradation of their enzymes, membranes, and organelles? Does controlling biological time influence cytoskeletal organization and cellular architecture in ways that confer evolutionary and physiological advantages? Potential answers to these fundamental questions of cell biology have historically revolved around the discussion of 'master' temporal programs, such as the principal cyclin-dependent kinase/cyclin cell division oscillator and the circadian clock. In this review, we provide an overview of the recent evidence supporting an emerging concept of 'autonomous clocks,' which under normal conditions can be entrained by the cell cycle and/or the circadian clock to run at their pace, but can also run independently to serve their functions if/when these major temporal programs are halted/abrupted. We begin the discussion by introducing recent developments in the study of such clocks and their roles at different scales and complexities. We then use current advances to elucidate the logic and molecular architecture of temporal networks that comprise autonomous clocks, providing important clues as to how these clocks may have evolved to run independently and, sometimes at the cost of redundancy, have strongly coupled to run under the full command of the cell cycle and/or the circadian clock. Next, we review a list of important recent findings that have shed new light onto potential hallmarks of autonomous clocks, suggestive of prospective theoretical and experimental approaches to further accelerate their discovery. Finally, we discuss their roles in health and disease, as well as possible therapeutic opportunities that targeting the autonomous clocks may offer.
Collapse
Affiliation(s)
- Mohammad Mofatteh
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Fabio Echegaray-Iturra
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Andrew Alamban
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Francesco Dalla Ricca
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Anand Bakshi
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Mustafa G Aydogan
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
28
|
Odermatt PD, Miettinen TP, Lemière J, Kang JH, Bostan E, Manalis SR, Huang KC, Chang F. Variations of intracellular density during the cell cycle arise from tip-growth regulation in fission yeast. eLife 2021; 10:64901. [PMID: 34100714 PMCID: PMC8221806 DOI: 10.7554/elife.64901] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/07/2021] [Indexed: 12/24/2022] Open
Abstract
Intracellular density impacts the physical nature of the cytoplasm and can globally affect cellular processes, yet density regulation remains poorly understood. Here, using a new quantitative phase imaging method, we determined that dry-mass density in fission yeast is maintained in a narrow distribution and exhibits homeostatic behavior. However, density varied during the cell cycle, decreasing during G2, increasing in mitosis and cytokinesis, and dropping rapidly at cell birth. These density variations were explained by a constant rate of biomass synthesis, coupled to slowdown of volume growth during cell division and rapid expansion post-cytokinesis. Arrest at specific cell-cycle stages exacerbated density changes. Spatially heterogeneous patterns of density suggested links between density regulation, tip growth, and intracellular osmotic pressure. Our results demonstrate that systematic density variations during the cell cycle are predominantly due to modulation of volume expansion, and reveal functional consequences of density gradients and cell-cycle arrests.
Collapse
Affiliation(s)
- Pascal D Odermatt
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States.,Department of Bioengineering, Stanford University, Stanford, United States
| | - Teemu P Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States.,MRC Laboratory for Molecular Cell Biology, University College, London, United Kingdom
| | - Joël Lemière
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Joon Ho Kang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States.,Department of Physics, Massachusetts Institute of Technology, Cambridge, United States.,Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Emrah Bostan
- Informatics Institute, University of Amsterdam, Amsterdamn, Netherlands
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, United States.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, United States.,Chan Zuckerberg Biohub, San Francisco, United States
| | - Fred Chang
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|