151
|
Matson JP, Cook JG. Cell cycle proliferation decisions: the impact of single cell analyses. FEBS J 2017; 284:362-375. [PMID: 27634578 PMCID: PMC5296213 DOI: 10.1111/febs.13898] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/23/2016] [Accepted: 09/13/2016] [Indexed: 12/16/2022]
Abstract
Cell proliferation is a fundamental requirement for organismal development and homeostasis. The mammalian cell division cycle is tightly controlled to ensure complete and precise genome duplication and segregation of replicated chromosomes to daughter cells. The onset of DNA replication marks an irreversible commitment to cell division, and the accumulated efforts of many decades of molecular and cellular studies have probed this cellular decision, commonly called the restriction point. Despite a long-standing conceptual framework of the restriction point for progression through G1 phase into S phase or exit from G1 phase to quiescence (G0), recent technical advances in quantitative single cell analysis of mammalian cells have provided new insights. Significant intercellular heterogeneity revealed by single cell studies and the discovery of discrete subpopulations in proliferating cultures suggests the need for an even more nuanced understanding of cell proliferation decisions. In this review, we describe some of the recent developments in the cell cycle field made possible by quantitative single cell experimental approaches.
Collapse
Affiliation(s)
- Jacob P. Matson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
| | - Jeanette G. Cook
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
| |
Collapse
|
152
|
Mahamed D, Boulle M, Ganga Y, Mc Arthur C, Skroch S, Oom L, Catinas O, Pillay K, Naicker M, Rampersad S, Mathonsi C, Hunter J, Wong EB, Suleman M, Sreejit G, Pym AS, Lustig G, Sigal A. Intracellular growth of Mycobacterium tuberculosis after macrophage cell death leads to serial killing of host cells. eLife 2017; 6. [PMID: 28130921 PMCID: PMC5319838 DOI: 10.7554/elife.22028] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 01/27/2017] [Indexed: 01/09/2023] Open
Abstract
A hallmark of pulmonary tuberculosis is the formation of macrophage-rich granulomas. These may restrict Mycobacterium tuberculosis (Mtb) growth, or progress to central necrosis and cavitation, facilitating pathogen growth. To determine factors leading to Mtb proliferation and host cell death, we used live cell imaging to track Mtb infection outcomes in individual primary human macrophages. Internalization of Mtb aggregates caused macrophage death, and phagocytosis of large aggregates was more cytotoxic than multiple small aggregates containing similar numbers of bacilli. Macrophage death did not result in clearance of Mtb. Rather, it led to accelerated intracellular Mtb growth regardless of prior activation or macrophage type. In contrast, bacillary replication was controlled in live phagocytes. Mtb grew as a clump in dead cells, and macrophages which internalized dead infected cells were very likely to die themselves, leading to a cell death cascade. This demonstrates how pathogen virulence can be achieved through numbers and aggregation states. DOI:http://dx.doi.org/10.7554/eLife.22028.001 Every year, around two million people worldwide die from tuberculosis, a disease caused by the bacterium Mycobacterium tuberculosis (Mtb). The bacteria generally infect the lungs. In response, the immune system forms structures called granulomas that attempt to control and isolate the infecting pathogens. Granulomas consist of immune cells known as macrophages, which engulf the M. tuberculosis bacteria and isolate them in a cellular compartment where the bacteria either cannot grow or are killed. However, if a large number of macrophages in a granuloma die, the granuloma’s core liquefies and the structure is coughed up into the airways, from where M. tuberculosis bacteria are transmitted to other people. But how do the bacteria manage to cause the extensive death of the cells that are supposed to control the infection? By imaging M. tuberculosis in human macrophages using time-lapse microscopy, Mahamed et al. reveal that the bacteria break down macrophage control by serially killing macrophages. M. tuberculosis cells first clump together and ‘gang up’ on a macrophage, which engulfs the clump and dies because the bacteria overwhelm it. This does not kill the bacteria, and they rapidly grow inside the dead macrophage. The dead cell is then cleaned up by another macrophage. However, the increasing number of bacteria inside the dead macrophage means that the new macrophage is even more likely to die than the first one. Hence, the bacteria use dead macrophages as fuel to grow on and as bait to attract the next immune cell. Overall, Mahamed et al. show that once a clump of M. tuberculosis initiates death of a single macrophage, it may lead to serial killing of other macrophages and a loss of control over the infection. An important next step will be to understand how the initial clump of bacteria is allowed to form. DOI:http://dx.doi.org/10.7554/eLife.22028.002
Collapse
Affiliation(s)
- Deeqa Mahamed
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Mikael Boulle
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa.,Max Planck Institute for Infection Biology, Berlin, Germany
| | - Yashica Ganga
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa
| | - Chanelle Mc Arthur
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Steven Skroch
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Lance Oom
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Oana Catinas
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa
| | - Kelly Pillay
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Myshnee Naicker
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa
| | - Sanisha Rampersad
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Colisile Mathonsi
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Jessica Hunter
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa
| | - Emily B Wong
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,Division of Infectious Diseases, Massachusetts General Hospital, Boston, United States
| | - Moosa Suleman
- Department of Pulmonology and Critical Care, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.,Department of Pulmonology, Inkosi Albert Luthuli Central Hospital, Durban, South Africa
| | | | - Alexander S Pym
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa
| | - Gila Lustig
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa
| | - Alex Sigal
- KwaZulu-Natal Research Institute for TB-HIV, Durban, South Africa.,University of KwaZulu-Natal, Durban, South Africa.,Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
153
|
Bergman HM, Lanekoff I. Profiling and quantifying endogenous molecules in single cells using nano-DESI MS. Analyst 2017; 142:3639-3647. [DOI: 10.1039/c7an00885f] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nano-DESI MS enables sensitive molecular profiling and quantification of endogenous species in single cells in a higher throughput manner.
Collapse
|
154
|
Ortega-Ferrusola C, Anel-López L, Martín-Muñoz P, Ortíz-Rodríguez JM, Gil MC, Alvarez M, de Paz P, Ezquerra LJ, Masot AJ, Redondo E, Anel L, Peña FJ. Computational flow cytometry reveals that cryopreservation induces spermptosis but subpopulations of spermatozoa may experience capacitation-like changes. Reproduction 2016; 153:293-304. [PMID: 27965398 DOI: 10.1530/rep-16-0539] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/02/2016] [Accepted: 12/12/2016] [Indexed: 12/19/2022]
Abstract
The reduced lifespan of cryopreserved spermatozoa in the mare reproductive tract has been attributed to both capacitative and apoptotic changes. However, there is a lack of studies investigating both phenomena simultaneously. In order to improve our knowledge in this particular point, we studied in raw and frozen-thawed samples apoptotic and capacitative markers using a wide battery of test based in flow cytometry. Apoptotic markers evaluated were caspase 3 activity, externalization of phosphatidylserine (PS), and mitochondrial membrane potential. Markers of changes resembling capacitation were membrane fluidity, tyrosine phosphorylation, and intracellular sodium. Conventional and computational flow cytometry using nonlinear dimensionally reduction techniques (t-distributed stochastic neighbor embedding (t-SNE)) and automatic classification of cellular expression by nonlinear stochastic embedding (ACCENSE) were used. Most of the changes induced by cryopreservation were apoptotic, with increase in caspase 3 activation (P < 0.01), PS translocation to the outer membrane (P < 0.001), loss of mitochondrial membrane potential (P < 0.05), and increase in intracellular Na+ (P < 0.01). Average values of markers of capacitative changes were not affected by cryopreservation; however, the analysis of the phenotype of individual spermatozoa using computational flow cytometry revealed the presence of subpopulations of spermatozoa experiencing capacitative changes. For the first time advanced computational techniques were applied to the analysis of spermatozoa, and these techniques were able to disclose relevant information of the ejaculate that remained hidden using conventional flow cytometry.
Collapse
Affiliation(s)
| | - L Anel-López
- Reproduction and Obstetrics Department of Animal Medicine and Surgery
| | - P Martín-Muñoz
- Laboratory of Equine Reproduction and Equine SpermatologyVeterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - J M Ortíz-Rodríguez
- Laboratory of Equine Reproduction and Equine SpermatologyVeterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - M C Gil
- Laboratory of Equine Reproduction and Equine SpermatologyVeterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - M Alvarez
- Reproduction and Obstetrics Department of Animal Medicine and Surgery
| | - P de Paz
- Department of Molecular BiologyUniversity of León, León, Spain
| | - L J Ezquerra
- Laboratory of Equine Reproduction and Equine SpermatologyVeterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - A J Masot
- Laboratory of Equine Reproduction and Equine SpermatologyVeterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - E Redondo
- Laboratory of Equine Reproduction and Equine SpermatologyVeterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - L Anel
- Reproduction and Obstetrics Department of Animal Medicine and Surgery
| | - F J Peña
- Laboratory of Equine Reproduction and Equine SpermatologyVeterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| |
Collapse
|
155
|
Modeling Cellular Noise Underlying Heterogeneous Cell Responses in the Epidermal Growth Factor Signaling Pathway. PLoS Comput Biol 2016; 12:e1005222. [PMID: 27902699 PMCID: PMC5130170 DOI: 10.1371/journal.pcbi.1005222] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/25/2016] [Indexed: 12/03/2022] Open
Abstract
Cellular heterogeneity, which plays an essential role in biological phenomena, such as drug resistance and migration, is considered to arise from intrinsic (i.e., reaction kinetics) and extrinsic (i.e., protein variability) noise in the cell. However, the mechanistic effects of these types of noise to determine the heterogeneity of signal responses have not been elucidated. Here, we report that the output of epidermal growth factor (EGF) signaling activity is modulated by cellular noise, particularly by extrinsic noise of particular signaling components in the pathway. We developed a mathematical model of the EGF signaling pathway incorporating regulation between extracellular signal-regulated kinase (ERK) and nuclear pore complex (NPC), which is necessary for switch-like activation of the nuclear ERK response. As the threshold of switch-like behavior is more sensitive to perturbations than the graded response, the effect of biological noise is potentially critical for cell fate decision. Our simulation analysis indicated that extrinsic noise, but not intrinsic noise, contributes to cell-to-cell heterogeneity of nuclear ERK. In addition, we accurately estimated variations in abundance of the signal proteins between individual cells by direct comparison of experimental data with simulation results using Apparent Measurement Error (AME). AME was constant regardless of whether the protein levels varied in a correlated manner, while covariation among proteins influenced cell-to-cell heterogeneity of nuclear ERK, suppressing the variation. Simulations using the estimated protein abundances showed that each protein species has different effects on cell-to-cell variation in the nuclear ERK response. In particular, variability of EGF receptor, Ras, Raf, and MEK strongly influenced cellular heterogeneity, while others did not. Overall, our results indicated that cellular heterogeneity in response to EGF is strongly driven by extrinsic noise, and that such heterogeneity results from variability of particular protein species that function as sensitive nodes, which may contribute to the pathogenesis of human diseases. Individual cell behaviors are controlled by a variety of noise, such as fluctuations in biochemical reactions, protein variability, molecular diffusion, transcriptional noise, cell-to-cell contact, temperature, and pH. Such cellular noise often interferes with signal responses from external stimuli, and such heterogeneity functions in induction of drug resistance, survival, and migration of cells. Thus, heterogeneous cellular responses have positive and negative roles. However, the regulatory mechanisms that produce cellular heterogeneity are unclear. By mathematical modeling and simulations, we investigated how heterogeneous signaling responses are evoked in the EGF signaling pathway and influence the switch-like activation of nuclear ERK. This study demonstrated that cellular heterogeneity of the EGF signaling response is evoked by cell-to-cell variation of particular signaling proteins, such as EGFR, Ras, Raf, and MEK, which act as sensitive nodes in the pathway. These results suggest that signaling responses in individual cells can be predicted from the levels of proteins of sensitive nodes. This study also suggested that proteins of sensitive nodes may serve as cell survival mechanisms.
Collapse
|
156
|
Digital Quantification of Proteins and mRNA in Single Mammalian Cells. Mol Cell 2016; 61:914-24. [PMID: 26990994 DOI: 10.1016/j.molcel.2016.02.030] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/19/2016] [Accepted: 02/25/2016] [Indexed: 01/23/2023]
Abstract
Absolute quantification of macromolecules in single cells is critical for understanding and modeling biological systems that feature cellular heterogeneity. Here we show extremely sensitive and absolute quantification of both proteins and mRNA in single mammalian cells by a very practical workflow that combines proximity ligation assay (PLA) and digital PCR. This digital PLA method has femtomolar sensitivity, which enables the quantification of very small protein concentration changes over its entire 3-log dynamic range, a quality necessary for accounting for single-cell heterogeneity. We counted both endogenous (CD147) and exogenously expressed (GFP-p65) proteins from hundreds of single cells and determined the correlation between CD147 mRNA and the protein it encodes. Using our data, a stochastic two-state model of the central dogma was constructed and verified using joint mRNA/protein distributions, allowing us to estimate transcription burst sizes and extrinsic noise strength and calculate the transcription and translation rate constants in single mammalian cells.
Collapse
|
157
|
Adamson A, Boddington C, Downton P, Rowe W, Bagnall J, Lam C, Maya-Mendoza A, Schmidt L, Harper CV, Spiller DG, Rand DA, Jackson DA, White MRH, Paszek P. Signal transduction controls heterogeneous NF-κB dynamics and target gene expression through cytokine-specific refractory states. Nat Commun 2016; 7:12057. [PMID: 27381163 PMCID: PMC4935804 DOI: 10.1038/ncomms12057] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 05/25/2016] [Indexed: 02/03/2023] Open
Abstract
Cells respond dynamically to pulsatile cytokine stimulation. Here we report that single, or well-spaced pulses of TNFα (>100 min apart) give a high probability of NF-κB activation. However, fewer cells respond to shorter pulse intervals (<100 min) suggesting a heterogeneous refractory state. This refractory state is established in the signal transduction network downstream of TNFR and upstream of IKK, and depends on the level of the NF-κB system negative feedback protein A20. If a second pulse within the refractory phase is IL-1β instead of TNFα, all of the cells respond. This suggests a mechanism by which two cytokines can synergistically activate an inflammatory response. Gene expression analyses show strong correlation between the cellular dynamic response and NF-κB-dependent target gene activation. These data suggest that refractory states in the NF-κB system constitute an inherent design motif of the inflammatory response and we suggest that this may avoid harmful homogenous cellular activation.
Collapse
Affiliation(s)
- Antony Adamson
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Christopher Boddington
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Polly Downton
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - William Rowe
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - James Bagnall
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Connie Lam
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Apolinar Maya-Mendoza
- The Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Lorraine Schmidt
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Claire V. Harper
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - David G. Spiller
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - David A. Rand
- Warwick Systems Biology and Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK
| | - Dean A. Jackson
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Michael R. H. White
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Pawel Paszek
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
158
|
Deep Learning in Label-free Cell Classification. Sci Rep 2016; 6:21471. [PMID: 26975219 PMCID: PMC4791545 DOI: 10.1038/srep21471] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/25/2016] [Indexed: 01/11/2023] Open
Abstract
Label-free cell analysis is essential to personalized genomics, cancer diagnostics, and drug development as it avoids adverse effects of staining reagents on cellular viability and cell signaling. However, currently available label-free cell assays mostly rely only on a single feature and lack sufficient differentiation. Also, the sample size analyzed by these assays is limited due to their low throughput. Here, we integrate feature extraction and deep learning with high-throughput quantitative imaging enabled by photonic time stretch, achieving record high accuracy in label-free cell classification. Our system captures quantitative optical phase and intensity images and extracts multiple biophysical features of individual cells. These biophysical measurements form a hyperdimensional feature space in which supervised learning is performed for cell classification. We compare various learning algorithms including artificial neural network, support vector machine, logistic regression, and a novel deep learning pipeline, which adopts global optimization of receiver operating characteristics. As a validation of the enhanced sensitivity and specificity of our system, we show classification of white blood T-cells against colon cancer cells, as well as lipid accumulating algal strains for biofuel production. This system opens up a new path to data-driven phenotypic diagnosis and better understanding of the heterogeneous gene expressions in cells.
Collapse
|
159
|
Dynamics inside the cancer cell attractor reveal cell heterogeneity, limits of stability, and escape. Proc Natl Acad Sci U S A 2016; 113:2672-7. [PMID: 26929366 DOI: 10.1073/pnas.1519210113] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The observed intercellular heterogeneity within a clonal cell population can be mapped as dynamical states clustered around an attractor point in gene expression space, owing to a balance between homeostatic forces and stochastic fluctuations. These dynamics have led to the cancer cell attractor conceptual model, with implications for both carcinogenesis and new therapeutic concepts. Immortalized and malignant EBV-carrying B-cell lines were used to explore this model and characterize the detailed structure of cell attractors. Any subpopulation selected from a population of cells repopulated the whole original basin of attraction within days to weeks. Cells at the basin edges were unstable and prone to apoptosis. Cells continuously changed states within their own attractor, thus driving the repopulation, as shown by fluorescent dye tracing. Perturbations of key regulatory genes induced a jump to a nearby attractor. Using the Fokker-Planck equation, this cell population behavior could be described as two virtual, opposing influences on the cells: one attracting toward the center and the other promoting diffusion in state space (noise). Transcriptome analysis suggests that these forces result from high-dimensional dynamics of the gene regulatory network. We propose that they can be generalized to all cancer cell populations and represent intrinsic behaviors of tumors, offering a previously unidentified characteristic for studying cancer.
Collapse
|
160
|
Webb AB, Lengyel IM, Jörg DJ, Valentin G, Jülicher F, Morelli LG, Oates AC. Persistence, period and precision of autonomous cellular oscillators from the zebrafish segmentation clock. eLife 2016; 5. [PMID: 26880542 PMCID: PMC4803185 DOI: 10.7554/elife.08438] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 02/11/2016] [Indexed: 12/11/2022] Open
Abstract
In vertebrate development, the sequential and rhythmic segmentation of the body axis
is regulated by a “segmentation clock”. This clock is comprised of a population of
coordinated oscillating cells that together produce rhythmic gene expression patterns
in the embryo. Whether individual cells autonomously maintain oscillations, or
whether oscillations depend on signals from neighboring cells is unknown. Using a
transgenic zebrafish reporter line for the cyclic transcription factor Her1, we
recorded single tailbud cells in vitro. We demonstrate that individual cells can
behave as autonomous cellular oscillators. We described the observed variability in
cell behavior using a theory of generic oscillators with correlated noise. Single
cells have longer periods and lower precision than the tissue, highlighting the role
of collective processes in the segmentation clock. Our work reveals a population of
cells from the zebrafish segmentation clock that behave as self-sustained, autonomous
oscillators with distinctive noisy dynamics. DOI:http://dx.doi.org/10.7554/eLife.08438.001 The timing and pattern of gene activity in cells can be very important. For example,
precise gene activity patterns in 24-hour circadian clocks help to set daily cycles
of rest and activity in organisms. In such scenarios, cells often communicate with
each other to coordinate the activity of their genes. To fully understand how the
behavior of the population emerges, scientists must first understand the gene
activity patterns in individual cells. Rhythmic gene activity is essential for the spinal column to form in fish and other
vertebrate embryos. A group of cells that switch genes on/off in a coordinated
pattern act like a clock to regulate the timing of the various steps in the process
of backbone formation. However, it is not clear if each cell is able to maintain a
rhythm of gene expression on their own, or whether they rely on messages from
neighboring cells to achieve it. Now, Webb et al. use time-lapse videos of individual cells isolated from the tail of
zebrafish embryos to show that each cell can maintain a pattern of rhythmic activity
in a gene called Her1. In the experiments, individual cells were
removed from zebrafish and placed under a microscope to record and track the activity
of Her1 over time using fluorescent proteins. These experiments show
that each cell is able to maintain a rhythmic pattern of Her1
expression on its own. Webb et al. then compared the Her1 activity patterns in individual
cells with the Her1 patterns present in a larger piece of zebrafish
tissue. The experiments showed that the rhythms in the individual cells are slower
and less precise in their timing than in the tissue. This suggests that groups of
cells must work together to create the synchronized rhythms of gene expression with
the right precision and timing needed for the spinal column to be patterned
correctly. In the future, further experiment with these cells will allow researchers to
investigate the genetic basis of the rhythms in single cells, and find out how
individual cells work together with their neighbors to allow tissues to work
properly. DOI:http://dx.doi.org/10.7554/eLife.08438.002
Collapse
Affiliation(s)
- Alexis B Webb
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Iván M Lengyel
- Departamento de Física, FCEyN UBA and IFIBA, CONICET, Buenos Aires, Argentina
| | - David J Jörg
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Guillaume Valentin
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Luis G Morelli
- Departamento de Física, FCEyN UBA and IFIBA, CONICET, Buenos Aires, Argentina
| | - Andrew C Oates
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
161
|
Computational Analysis of AMPK-Mediated Neuroprotection Suggests Acute Excitotoxic Bioenergetics and Glucose Dynamics Are Regulated by a Minimal Set of Critical Reactions. PLoS One 2016; 11:e0148326. [PMID: 26840769 PMCID: PMC4740490 DOI: 10.1371/journal.pone.0148326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/15/2016] [Indexed: 11/22/2022] Open
Abstract
Loss of ionic homeostasis during excitotoxic stress depletes ATP levels and activates the AMP-activated protein kinase (AMPK), re-establishing energy production by increased expression of glucose transporters on the plasma membrane. Here, we develop a computational model to test whether this AMPK-mediated glucose import can rapidly restore ATP levels following a transient excitotoxic insult. We demonstrate that a highly compact model, comprising a minimal set of critical reactions, can closely resemble the rapid dynamics and cell-to-cell heterogeneity of ATP levels and AMPK activity, as confirmed by single-cell fluorescence microscopy in rat primary cerebellar neurons exposed to glutamate excitotoxicity. The model further correctly predicted an excitotoxicity-induced elevation of intracellular glucose, and well resembled the delayed recovery and cell-to-cell heterogeneity of experimentally measured glucose dynamics. The model also predicted necrotic bioenergetic collapse and altered calcium dynamics following more severe excitotoxic insults. In conclusion, our data suggest that a minimal set of critical reactions may determine the acute bioenergetic response to transient excitotoxicity and that an AMPK-mediated increase in intracellular glucose may be sufficient to rapidly recover ATP levels following an excitotoxic insult.
Collapse
|
162
|
Venturelli OS, Egbert RG, Arkin AP. Towards Engineering Biological Systems in a Broader Context. J Mol Biol 2016; 428:928-44. [DOI: 10.1016/j.jmb.2015.10.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/24/2015] [Accepted: 10/28/2015] [Indexed: 01/18/2023]
|
163
|
Ferraro T, Esposito E, Mancini L, Ng S, Lucas T, Coppey M, Dostatni N, Walczak AM, Levine M, Lagha M. Transcriptional Memory in the Drosophila Embryo. Curr Biol 2016; 26:212-218. [PMID: 26748851 PMCID: PMC4970865 DOI: 10.1016/j.cub.2015.11.058] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/05/2015] [Accepted: 11/12/2015] [Indexed: 02/04/2023]
Abstract
Transmission of active transcriptional states from mother to daughter cells has the potential to foster precision in the gene expression programs underlying development. Such transcriptional memory has been specifically proposed to promote rapid reactivation of complex gene expression profiles after successive mitoses in Drosophila development [1]. By monitoring transcription in living Drosophila embryos, we provide the first evidence for transcriptional memory in animal development. We specifically monitored the activities of stochastically expressed transgenes in order to distinguish active and inactive mother cells and the behaviors of their daughter nuclei after mitosis. Quantitative analyses reveal that there is a 4-fold higher probability for rapid reactivation after mitosis when the mother experienced transcription. Moreover, memory nuclei activate transcription twice as fast as neighboring inactive mothers, thus leading to augmented levels of gene expression. We propose that transcriptional memory is a mechanism of precision, which helps coordinate gene activity during embryogenesis.
Collapse
Affiliation(s)
- Teresa Ferraro
- Institut Curie, PSL Research University, UMR 3664/UMR 168, Paris 75248, France; CNRS, UMR 3664/UMR 168/UMR 8549/UMR 8550, Paris 75248, France; Sorbonne Universités, UPMC University Paris 06, UMR 3664/UMR 168, Paris 75248, France; PSL, Ecole Normale Supérieure, UMR 8549, Paris 75005, France
| | - Emilia Esposito
- Molecular and Cellular Biology Department, GDD, University of California, Berkeley, Berkeley, CA 94720, USA; Lewis-Sigler Institute, Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Laure Mancini
- Molecular and Cellular Biology Department, GDD, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sam Ng
- Molecular and Cellular Biology Department, GDD, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Tanguy Lucas
- Institut Curie, PSL Research University, UMR 3664/UMR 168, Paris 75248, France; CNRS, UMR 3664/UMR 168/UMR 8549/UMR 8550, Paris 75248, France; Sorbonne Universités, UPMC University Paris 06, UMR 3664/UMR 168, Paris 75248, France
| | - Mathieu Coppey
- Institut Curie, PSL Research University, UMR 3664/UMR 168, Paris 75248, France; CNRS, UMR 3664/UMR 168/UMR 8549/UMR 8550, Paris 75248, France; Sorbonne Universités, UPMC University Paris 06, UMR 3664/UMR 168, Paris 75248, France
| | - Nathalie Dostatni
- Institut Curie, PSL Research University, UMR 3664/UMR 168, Paris 75248, France; CNRS, UMR 3664/UMR 168/UMR 8549/UMR 8550, Paris 75248, France; Sorbonne Universités, UPMC University Paris 06, UMR 3664/UMR 168, Paris 75248, France
| | - Aleksandra M Walczak
- CNRS, UMR 3664/UMR 168/UMR 8549/UMR 8550, Paris 75248, France; PSL, Ecole Normale Supérieure, UMR 8549, Paris 75005, France
| | - Michael Levine
- Molecular and Cellular Biology Department, GDD, University of California, Berkeley, Berkeley, CA 94720, USA; Lewis-Sigler Institute, Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Mounia Lagha
- Molecular and Cellular Biology Department, GDD, University of California, Berkeley, Berkeley, CA 94720, USA; IGMM, CNRS, UMR 5535, Montpellier 34293, France.
| |
Collapse
|
164
|
Kume K, Ishida K, Ikeda M, Takemoto K, Shimura T, Young L, Nishizuka SS. Systematic Protein Level Regulation via Degradation Machinery Induced by Genotoxic Drugs. J Proteome Res 2016; 15:205-15. [PMID: 26625007 DOI: 10.1021/acs.jproteome.5b00759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In this study we monitored protein dynamics in response to cisplatin, 5-fluorouracil, and irinotecan with different concentrations and administration modes using "reverse-phase" protein arrays (RPPAs) in order to gain comprehensive insight into the protein dynamics induced by genotoxic drugs. Among 666 protein time-courses, 38% exhibited an increasing trend, 32% exhibited a steady decrease, and 30% fluctuated within 24 h after drug exposure. We analyzed almost 12,000 time-course pairs of protein levels based on the geometrical similarity by correlation distance (dCor). Twenty-two percent of the pairs showed dCor > 0.8, which indicates that each protein of the pair had similar dynamics. These trends were disrupted by a proteasome inhibitor, MG132, suggesting that the protein degradation system was activated in response to the drugs. Among the pairs with high dCor, the average dCor of pairs with apoptosis-related protein was significantly higher than those without, indicating that regulation of protein levels was induced by the drugs. These results suggest that the levels of numerous functionally distinct proteins may be regulated by common degradation machinery induced by genotoxic drugs.
Collapse
Affiliation(s)
- Kohei Kume
- Medical Innovation for Advanced Science and Technology program (MIAST), Iwate Medical University , Morioka, Iwate 020-8505, Japan.,Institute for Biomedical Sciences, Iwate Medical University , Yahaba, Iwate 020-8505, Japan
| | | | | | - Kazuhiro Takemoto
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology , Iizuka, Fukuoka 820-8502, Japan
| | - Tsutomu Shimura
- Department of Environmental Health, National Institute of Public Health , Wako-shi, Saitama 351-097, Japan
| | - Lynn Young
- National Institutes of Health (NIH) Library, Division of Library Services, Office of Research Services, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Satoshi S Nishizuka
- Medical Innovation for Advanced Science and Technology program (MIAST), Iwate Medical University , Morioka, Iwate 020-8505, Japan.,Institute for Biomedical Sciences, Iwate Medical University , Yahaba, Iwate 020-8505, Japan.,Department of Surgery, Iwate Medical University School of Dentistry , Morioka, Iwate 020-8505, Japan
| |
Collapse
|
165
|
Mackey MC, Tyran-Kamińska M. The limiting dynamics of a bistable molecular switch with and without noise. J Math Biol 2015; 73:367-95. [PMID: 26692266 DOI: 10.1007/s00285-015-0949-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 08/23/2015] [Indexed: 11/26/2022]
Abstract
We consider the dynamics of a population of organisms containing two mutually inhibitory gene regulatory networks, that can result in a bistable switch-like behaviour. We completely characterize their local and global dynamics in the absence of any noise, and then go on to consider the effects of either noise coming from bursting (transcription or translation), or Gaussian noise in molecular degradation rates when there is a dominant slow variable in the system. We show analytically how the steady state distribution in the population can range from a single unimodal distribution through a bimodal distribution and give the explicit analytic form for the invariant stationary density which is globally asymptotically stable. Rather remarkably, the behaviour of the stationary density with respect to the parameters characterizing the molecular behaviour of the bistable switch is qualitatively identical in the presence of noise coming from bursting as well as in the presence of Gaussian noise in the degradation rate. This implies that one cannot distinguish between either the dominant source or nature of noise based on the stationary molecular distribution in a population of cells. We finally show that the switch model with bursting but two dominant slow genes has an asymptotically stable stationary density.
Collapse
Affiliation(s)
- Michael C Mackey
- Departments of Physiology, Physics and Mathematics, Centre for Applied Mathematics in Bioscience and Medicine (CAMBAM), McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada.
| | - Marta Tyran-Kamińska
- Institute of Mathematics, University of Silesia, Bankowa 14, 40-007, Katowice, Poland
| |
Collapse
|
166
|
Jenkins RP, Hanisch A, Soza-Ried C, Sahai E, Lewis J. Stochastic Regulation of her1/7 Gene Expression Is the Source of Noise in the Zebrafish Somite Clock Counteracted by Notch Signalling. PLoS Comput Biol 2015; 11:e1004459. [PMID: 26588097 PMCID: PMC4654481 DOI: 10.1371/journal.pcbi.1004459] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 07/09/2015] [Indexed: 12/30/2022] Open
Abstract
The somite segmentation clock is a robust oscillator used to generate regularly-sized segments during early vertebrate embryogenesis. It has been proposed that the clocks of neighbouring cells are synchronised via inter-cellular Notch signalling, in order to overcome the effects of noisy gene expression. When Notch-dependent communication between cells fails, the clocks of individual cells operate erratically and lose synchrony over a period of about 5 to 8 segmentation clock cycles (2-3 hours in the zebrafish). Here, we quantitatively investigate the effects of stochasticity on cell synchrony, using mathematical modelling, to investigate the likely source of such noise. We find that variations in the transcription, translation and degradation rate of key Notch signalling regulators do not explain the in vivo kinetics of desynchronisation. Rather, the analysis predicts that clock desynchronisation, in the absence of Notch signalling, is due to the stochastic dissociation of Her1/7 repressor proteins from the oscillating her1/7 autorepressed target genes. Using in situ hybridisation to visualise sites of active her1 transcription, we measure an average delay of approximately three minutes between the times of activation of the two her1 alleles in a cell. Our model shows that such a delay is sufficient to explain the in vivo rate of clock desynchronisation in Notch pathway mutant embryos and also that Notch-mediated synchronisation is sufficient to overcome this stochastic variation. This suggests that the stochastic nature of repressor/DNA dissociation is the major source of noise in the segmentation clock.
Collapse
Affiliation(s)
- Robert P. Jenkins
- Tumour Cell Biology Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Anja Hanisch
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Cristian Soza-Ried
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Julian Lewis
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| |
Collapse
|
167
|
Skataric M, Nikolaev EV, Sontag ED. Fundamental limitation of the instantaneous approximation in fold-change detection models. IET Syst Biol 2015; 9:1-15. [PMID: 25569859 DOI: 10.1049/iet-syb.2014.0006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The phenomenon of fold-change detection, or scale-invariance, is exhibited by a variety of sensory systems, in both bacterial and eukaryotic signalling pathways. It has been often remarked in the systems biology literature that certain systems whose output variables respond at a faster time scale than internal components give rise to an approximate scale-invariant behaviour, allowing approximate fold-change detection in stimuli. This study establishes a fundamental limitation of such a mechanism, showing that there is a minimal fold-change detection error that cannot be overcome, no matter how large the separation of time scales is. To illustrate this theoretically predicted limitation, the authors discuss two common biomolecular network motifs, an incoherent feedforward loop and a feedback system, as well as a published model of the chemotaxis signalling pathway of Dictyostelium discoideum.
Collapse
Affiliation(s)
- Maja Skataric
- Department of Electrical and Computer Engineering, Rutgers University, Piscataway, NJ 08854-8019, USA
| | - Evgeni V Nikolaev
- Department of Mathematics, Rutgers University, Piscataway, NJ 08854-8019, USA
| | - Eduardo D Sontag
- Department of Mathematics, Rutgers University, Piscataway, NJ 08854-8019, USA.
| |
Collapse
|
168
|
Handly LN, Pilko A, Wollman R. Paracrine communication maximizes cellular response fidelity in wound signaling. eLife 2015; 4:e09652. [PMID: 26448485 PMCID: PMC4686426 DOI: 10.7554/elife.09652] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/07/2015] [Indexed: 11/15/2022] Open
Abstract
Population averaging due to paracrine communication can arbitrarily reduce cellular response variability. Yet, variability is ubiquitously observed, suggesting limits to paracrine averaging. It remains unclear whether and how biological systems may be affected by such limits of paracrine signaling. To address this question, we quantify the signal and noise of Ca(2+) and ERK spatial gradients in response to an in vitro wound within a novel microfluidics-based device. We find that while paracrine communication reduces gradient noise, it also reduces the gradient magnitude. Accordingly we predict the existence of a maximum gradient signal to noise ratio. Direct in vitro measurement of paracrine communication verifies these predictions and reveals that cells utilize optimal levels of paracrine signaling to maximize the accuracy of gradient-based positional information. Our results demonstrate the limits of population averaging and show the inherent tradeoff in utilizing paracrine communication to regulate cellular response fidelity.
Collapse
Affiliation(s)
- L Naomi Handly
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, United States
| | - Anna Pilko
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, United States
| | - Roy Wollman
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, United States
- Section for Cell and Developmental Biology , Division of Biological Sciences, University of California, San Diego, La Jolla, United States
- San Diego Center for Systems Biology, La Jolla, United States
| |
Collapse
|
169
|
Abstract
Microbes transiently differentiate into distinct, specialized cell types to generate functional diversity and cope with changing environmental conditions. Though alternate programs often entail radically different physiological and morphological states, recent single-cell studies have revealed that these crucial decisions are often left to chance. In these cases, the underlying genetic circuits leverage the intrinsic stochasticity of intracellular chemistry to drive transition between states. Understanding how these circuits transform transient gene expression fluctuations into lasting phenotypic programs will require a combination of quantitative modeling and extensive, time-resolved observation of switching events in single cells. In this article, we survey microbial cell fate decisions demonstrated to involve a random element, describe theoretical frameworks for understanding stochastic switching between states, and highlight recent advances in microfluidics that will enable characterization of key dynamic features of these circuits.
Collapse
Affiliation(s)
- Thomas M Norman
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115; , ,
| | - Nathan D Lord
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115; , ,
| | - Johan Paulsson
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115; , ,
| | - Richard Losick
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138;
| |
Collapse
|
170
|
|
171
|
Flusberg DA, Sorger PK. Surviving apoptosis: life-death signaling in single cells. Trends Cell Biol 2015; 25:446-58. [PMID: 25920803 PMCID: PMC4570028 DOI: 10.1016/j.tcb.2015.03.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/19/2015] [Accepted: 03/19/2015] [Indexed: 12/16/2022]
Abstract
Tissue development and homeostasis are regulated by opposing pro-survival and pro-death signals. An interesting feature of the Tumor Necrosis Factor (TNF) family of ligands is that they simultaneously activate opposing signals within a single cell via the same ligand-receptor complex. The magnitude of pro-death events such as caspase activation and pro-survival events such as Nuclear Factor (NF)-κB activation vary not only from one cell type to the next but also among individual cells of the same type due to intrinsic and extrinsic noise. The molecules involved in these pro-survival and/or pro-death pathways, and the different phenotypes that result from their activities, have been recently reviewed. Here we focus on the impact of cell-to-cell variability in the strength of these opposing signals on shaping cell fate decisions.
Collapse
Affiliation(s)
- Deborah A Flusberg
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
172
|
Cannon D, Corrigan AM, Miermont A, McDonel P, Chubb JR. Multiple cell and population-level interactions with mouse embryonic stem cell heterogeneity. Development 2015. [PMID: 26209649 DOI: 10.1242/dev.120741] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Much of development and disease concerns the generation of gene expression differences between related cells sharing similar niches. However, most analyses of gene expression only assess population and time-averaged levels of steady-state transcription. The mechanisms driving differentiation are buried within snapshots of the average cell, lacking dynamic information and the diverse regulatory history experienced by individual cells. Here, we use a quantitative imaging platform with large time series data sets to determine the regulation of developmental gene expression by cell cycle, lineage, motility and environment. We apply this technology to the regulation of the pluripotency gene Nanog in mouse embryonic stem cells. Our data reveal the diversity of cell and population-level interactions with Nanog dynamics and heterogeneity, and how this regulation responds to triggers of pluripotency. Cell cycles are highly heterogeneous and cycle time increases with Nanog reporter expression, with longer, more variable cycle times as cells approach ground-state pluripotency. Nanog reporter expression is highly stable over multiple cell generations, with fluctuations within cycles confined by an attractor state. Modelling reveals an environmental component to expression stability, in addition to any cell-autonomous behaviour, and we identify interactions of cell density with both cycle behaviour and Nanog. Rex1 expression dynamics showed shared and distinct regulatory effects. Overall, our observations of multiple partially overlapping dynamic heterogeneities imply complex cell and environmental regulation of pluripotent cell behaviour, and suggest simple deterministic views of stem cell states are inappropriate.
Collapse
Affiliation(s)
- Danielle Cannon
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Adam M Corrigan
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Agnes Miermont
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Patrick McDonel
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA 02142, USA
| | - Jonathan R Chubb
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
173
|
Lee U, Skinner JJ, Reinitz J, Rosner MR, Kim EJ. Noise-Driven Phenotypic Heterogeneity with Finite Correlation Time in Clonal Populations. PLoS One 2015; 10:e0132397. [PMID: 26203903 PMCID: PMC4512695 DOI: 10.1371/journal.pone.0132397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/12/2015] [Indexed: 11/19/2022] Open
Abstract
There has been increasing awareness in the wider biological community of the role of clonal phenotypic heterogeneity in playing key roles in phenomena such as cellular bet-hedging and decision making, as in the case of the phage-λ lysis/lysogeny and B. Subtilis competence/vegetative pathways. Here, we report on the effect of stochasticity in growth rate, cellular memory/intermittency, and its relation to phenotypic heterogeneity. We first present a linear stochastic differential model with finite auto-correlation time, where a randomly fluctuating growth rate with a negative average is shown to result in exponential growth for sufficiently large fluctuations in growth rate. We then present a non-linear stochastic self-regulation model where the loss of coherent self-regulation and an increase in noise can induce a shift from bounded to unbounded growth. An important consequence of these models is that while the average change in phenotype may not differ for various parameter sets, the variance of the resulting distributions may considerably change. This demonstrates the necessity of understanding the influence of variance and heterogeneity within seemingly identical clonal populations, while providing a mechanism for varying functional consequences of such heterogeneity. Our results highlight the importance of a paradigm shift from a deterministic to a probabilistic view of clonality in understanding selection as an optimization problem on noise-driven processes, resulting in a wide range of biological implications, from robustness to environmental stress to the development of drug resistance.
Collapse
Affiliation(s)
- UnJin Lee
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, United States of America
| | - John J. Skinner
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, United States of America
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, United States of America
| | - John Reinitz
- Departments of Statistics, Ecology and Evolution, Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, United States of America
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, United States of America
| | - Eun-Jin Kim
- School of Mathematics and Statistics, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
174
|
Roth S, Kholodenko BN, Smit MJ, Bruggeman FJ. G Protein-Coupled Receptor Signaling Networks from a Systems Perspective. Mol Pharmacol 2015; 88:604-16. [PMID: 26162865 DOI: 10.1124/mol.115.100057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/10/2015] [Indexed: 12/20/2022] Open
Abstract
The signal-transduction network of a mammalian cell integrates internal and external cues to initiate adaptive responses. Among the cell-surface receptors are the G protein-coupled receptors (GPCRs), which have remarkable signal-integrating capabilities. Binding of extracellular signals stabilizes intracellular-domain conformations that selectively activate intracellular proteins. Hereby, multiple signaling routes are activated simultaneously to degrees that are signal-combination dependent. Systems-biology studies indicate that signaling networks have emergent processing capabilities that go far beyond those of single proteins. Such networks are spatiotemporally organized and capable of gradual, oscillatory, all-or-none, and subpopulation-generating responses. Protein-protein interactions, generating feedback and feedforward circuitry, are generally required for these spatiotemporal phenomena. Understanding of information processing by signaling networks therefore requires network theories in addition to biochemical and biophysical concepts. Here we review some of the key signaling systems behaviors that have been discovered recurrently across signaling networks. We emphasize the role of GPCRs, so far underappreciated receptors in systems-biology research.
Collapse
Affiliation(s)
- S Roth
- Systems Bioinformatics (S.R., F.J.B.) and Amsterdam Institute for Molecules, Medicines & Systems, VU University, Amsterdam, The Netherlands (M.J.S.); and Systems Biology Ireland, University College Dublin, Dublin, Ireland (B.N.K.)
| | - B N Kholodenko
- Systems Bioinformatics (S.R., F.J.B.) and Amsterdam Institute for Molecules, Medicines & Systems, VU University, Amsterdam, The Netherlands (M.J.S.); and Systems Biology Ireland, University College Dublin, Dublin, Ireland (B.N.K.)
| | - M J Smit
- Systems Bioinformatics (S.R., F.J.B.) and Amsterdam Institute for Molecules, Medicines & Systems, VU University, Amsterdam, The Netherlands (M.J.S.); and Systems Biology Ireland, University College Dublin, Dublin, Ireland (B.N.K.)
| | - F J Bruggeman
- Systems Bioinformatics (S.R., F.J.B.) and Amsterdam Institute for Molecules, Medicines & Systems, VU University, Amsterdam, The Netherlands (M.J.S.); and Systems Biology Ireland, University College Dublin, Dublin, Ireland (B.N.K.)
| |
Collapse
|
175
|
Langereis MA, Bakkers MJG, Deng L, Padler-Karavani V, Vervoort SJ, Hulswit RJG, van Vliet ALW, Gerwig GJ, de Poot SAH, Boot W, van Ederen AM, Heesters BA, van der Loos CM, van Kuppeveld FJM, Yu H, Huizinga EG, Chen X, Varki A, Kamerling JP, de Groot RJ. Complexity and Diversity of the Mammalian Sialome Revealed by Nidovirus Virolectins. Cell Rep 2015; 11:1966-78. [PMID: 26095364 PMCID: PMC5292239 DOI: 10.1016/j.celrep.2015.05.044] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 05/01/2015] [Accepted: 05/22/2015] [Indexed: 12/23/2022] Open
Abstract
Sialic acids (Sias), 9-carbon-backbone sugars, are among the most complex and versatile molecules of life. As terminal residues of glycans on proteins and lipids, Sias are key elements of glycotopes of both cellular and microbial lectins and thus act as important molecular tags in cell recognition and signaling events. Their functions in such interactions can be regulated by post-synthetic modifications, the most common of which is differential Sia-O-acetylation (O-Ac-Sias). The biology of O-Ac-Sias remains mostly unexplored, largely because of limitations associated with their specific in situ detection. Here, we show that dual-function hemagglutinin-esterase envelope proteins of nidoviruses distinguish between a variety of closely related O-Ac-Sias. By using soluble forms of hemagglutinin-esterases as lectins and sialate-O-acetylesterases, we demonstrate differential expression of distinct O-Ac-sialoglycan populations in an organ-, tissue- and cell-specific fashion. Our findings indicate that programmed Sia-O-acetylation/de-O-acetylation may be critical to key aspects of cell development, homeostasis, and/or function. Virolectins detect and distinguish between closely related O-Ac-Sias in situ O-Ac-sialoglycans occur in nature in a diversity not appreciated so far O-Ac-Sias are differentially expressed in a species-, tissue-, and cell-specific fashion There is extensive cell-to-cell variability in O-Ac-Sia expression in vivo and in vitro
Collapse
Affiliation(s)
- Martijn A Langereis
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Mark J G Bakkers
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Lingquan Deng
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Vered Padler-Karavani
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Stephin J Vervoort
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Ruben J G Hulswit
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Arno L W van Vliet
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Gerrit J Gerwig
- Bio-Organic Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Stefanie A H de Poot
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Willemijn Boot
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Anne Marie van Ederen
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Balthasar A Heesters
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Chris M van der Loos
- Department of Cardiovascular Pathology, Free University Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Frank J M van Kuppeveld
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Hai Yu
- Department of Chemistry, University of California, Davis, Davis, CA 95616, USA
| | - Eric G Huizinga
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Xi Chen
- Department of Chemistry, University of California, Davis, Davis, CA 95616, USA
| | - Ajit Varki
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Johannis P Kamerling
- Bio-Organic Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Raoul J de Groot
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands.
| |
Collapse
|
176
|
Dexter JP, Dasgupta T, Gunawardena J. Invariants reveal multiple forms of robustness in bifunctional enzyme systems. Integr Biol (Camb) 2015; 7:883-94. [PMID: 26021467 DOI: 10.1039/c5ib00009b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Experimental and theoretical studies have suggested that bifunctional enzymes catalyzing opposing modification and demodification reactions can confer steady-state concentration robustness to their substrates. However, the types of robustness and the biochemical basis for them have remained elusive. Here we report a systematic study of the most general biochemical reaction network for a bifunctional enzyme acting on a substrate with one modification site, along with eleven sub-networks with more specialized biochemical assumptions. We exploit ideas from computational algebraic geometry, introduced in previous work, to find a polynomial expression (an invariant) between the steady state concentrations of the modified and unmodified substrate for each network. We use these invariants to identify five classes of robust behavior: robust upper bounds on concentration, robust two-sided bounds on concentration ratio, hybrid robustness, absolute concentration robustness (ACR), and robust concentration ratio. This analysis demonstrates that robustness can take a variety of forms and that the type of robustness is sensitive to many biochemical details, with small changes in biochemistry leading to very different steady-state behaviors. In particular, we find that the widely-studied ACR requires highly specialized assumptions in addition to bifunctionality. An unexpected result is that the robust bounds derived from invariants are strictly tighter than those derived by ad hoc manipulation of the underlying differential equations, confirming the value of invariants as a tool to gain insight into biochemical reaction networks. Furthermore, invariants yield multiple experimentally testable predictions and illuminate new strategies for inferring enzymatic mechanisms from steady-state measurements.
Collapse
Affiliation(s)
- Joseph P Dexter
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | | | | |
Collapse
|
177
|
Kiss A, Gong X, Kowalewski JM, Shafqat-Abbasi H, Strömblad S, Lock JG. Non-monotonic cellular responses to heterogeneity in talin protein expression-level. Integr Biol (Camb) 2015; 7:1171-85. [PMID: 26000342 DOI: 10.1039/c4ib00291a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Talin is a key cell-matrix adhesion component with a central role in regulating adhesion complex maturation, and thereby various cellular properties including adhesion and migration. However, knockdown studies have produced inconsistent findings regarding the functional influence of talin in these processes. Such discrepancies may reflect non-monotonic responses to talin expression-level variation that are not detectable via canonical "binary" comparisons of aggregated control versus knockdown cell populations. Here, we deployed an "analogue" approach to map talin influence across a continuous expression-level spectrum, which we extended with sub-maximal RNAi-mediated talin depletion. Applying correlative imaging to link live cell and fixed immunofluorescence data on a single cell basis, we related per cell talin levels to per cell measures quantitatively defining an array of cellular properties. This revealed both linear and non-linear correspondences between talin expression and cellular properties, including non-monotonic influences over cell shape, adhesion complex-F-actin association and adhesion localization. Furthermore, we demonstrate talin level-dependent changes in networks of correlations among adhesion/migration properties, particularly in relation to cell migration speed. Importantly, these correlation networks were strongly affected by talin expression heterogeneity within the natural range, implying that this endogenous variation has a broad, quantitatively detectable influence. Overall, we present an accessible analogue method that reveals complex dependencies on talin expression-level, thereby establishing a framework for considering non-linear and non-monotonic effects of protein expression-level heterogeneity in cellular systems.
Collapse
Affiliation(s)
- Alexa Kiss
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Hälsov. 7-9, G-building floor 6, S-141 83 Huddinge, Sweden.
| | | | | | | | | | | |
Collapse
|
178
|
Lee TJ, Wong J, Bae S, Lee AJ, Lopatkin A, Yuan F, You L. A power-law dependence of bacterial invasion on mammalian host receptors. PLoS Comput Biol 2015; 11:e1004203. [PMID: 25879937 PMCID: PMC4399907 DOI: 10.1371/journal.pcbi.1004203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/19/2015] [Indexed: 01/04/2023] Open
Abstract
Pathogenic bacteria such as Listeria and Yersinia gain initial entry by binding to host target cells and stimulating their internalization. Bacterial uptake entails successive, increasingly strong associations between receptors on the surface of bacteria and hosts. Even with genetically identical cells grown in the same environment, there are vast differences in the number of bacteria entering any given cell. To gain insight into this variability, we examined uptake dynamics of Escherichia coli engineered to express the invasin surface receptor from Yersinia, which enables uptake via mammalian host β1-integrins. Surprisingly, we found that the uptake probability of a single bacterium follows a simple power-law dependence on the concentration of integrins. Furthermore, the value of a power-law parameter depends on the particular host-bacterium pair but not on bacterial concentration. This power-law captures the complex, variable processes underlying bacterial invasion while also enabling differentiation of cell lines. Uptake of bacteria by mammalian cells is highly variable within a population of host cells and between host cell types. A detailed but unwieldy mechanistic model describing individual host-pathogen receptor binding events is captured by a simple power-law dependence on the concentration of the host receptors. The power-law parameters capture characteristics of the host-bacterium pair interaction and can differentiate host cell lines. This study has important implications for understanding the accuracy and precision of therapeutics employing receptor-mediated transport of materials to mammalian hosts.
Collapse
Affiliation(s)
- Tae J. Lee
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Jeffrey Wong
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Sena Bae
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Anna Jisu Lee
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Allison Lopatkin
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
- Center for Systems Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
179
|
Betts Z, Croxford AS, Dickson AJ. Evaluating the interaction between UCOE and DHFR-linked amplification and stability of recombinant protein expression. Biotechnol Prog 2015; 31:1014-25. [DOI: 10.1002/btpr.2083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/17/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Zeynep Betts
- Faculty of Life Sciences; University of Manchester; Michael Smith Building, Oxford Road Manchester M13 9PT UK
| | - Alexandra S Croxford
- Faculty of Life Sciences; University of Manchester; Michael Smith Building, Oxford Road Manchester M13 9PT UK
| | - Alan J Dickson
- Faculty of Life Sciences; University of Manchester; Michael Smith Building, Oxford Road Manchester M13 9PT UK
| |
Collapse
|
180
|
Sandler O, Mizrahi SP, Weiss N, Agam O, Simon I, Balaban NQ. Lineage correlations of single cell division time as a probe of cell-cycle dynamics. Nature 2015; 519:468-71. [DOI: 10.1038/nature14318] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 02/13/2015] [Indexed: 11/09/2022]
|
181
|
Schubert V, Weisshart K. Abundance and distribution of RNA polymerase II in Arabidopsis interphase nuclei. JOURNAL OF EXPERIMENTAL BOTANY 2015; 66:1687-98. [PMID: 25740920 PMCID: PMC4357323 DOI: 10.1093/jxb/erv091] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
RNA polymerase II (RNAPII) is responsible for the transcription of most eukaryotic protein-coding genes. Analysing the topological distribution and quantification of RNAPII can contribute to understanding its function in interphase nuclei. Previously it was shown that RNAPII molecules in plant nuclei form reticulate structures within euchromatin of differentiated Arabidopsis thaliana nuclei rather than being organized in distinct 'transcription factories' as observed in mammalian nuclei. Immunosignal intensity measurements based on specific antibody labelling in maximum intensity projections of image stacks acquired by structured illumination microscopy (SIM) suggested a relative proportional increase of RNAPII in endopolyploid plant nuclei. Here, photoactivated localization microscopy (PALM) was applied to determine the absolute number and distribution of active and inactive RNAPII molecules in differentiated A. thaliana nuclei. The proportional increase of RNAPII during endopolyploidization is confirmed, but it is also shown that PALM measurements are more reliable than those based on SIM in terms of quantification. The single molecule localization results show that, although RNAPII molecules are globally dispersed within plant euchromatin, they also aggregate within smaller distances as described for mammalian transcription factories.
Collapse
Affiliation(s)
- Veit Schubert
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, D-06466 Stadt Seeland, Germany
| | | |
Collapse
|
182
|
Chisholm RH, Lorenzi T, Lorz A, Larsen AK, de Almeida LN, Escargueil A, Clairambault J. Emergence of drug tolerance in cancer cell populations: an evolutionary outcome of selection, nongenetic instability, and stress-induced adaptation. Cancer Res 2015; 75:930-9. [PMID: 25627977 DOI: 10.1158/0008-5472.can-14-2103] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In recent experiments on isogenetic cancer cell lines, it was observed that exposure to high doses of anticancer drugs can induce the emergence of a subpopulation of weakly proliferative and drug-tolerant cells, which display markers associated with stem cell-like cancer cells. After a period of time, some of the surviving cells were observed to change their phenotype to resume normal proliferation and eventually repopulate the sample. Furthermore, the drug-tolerant cells could be drug resensitized following drug washout. Here, we propose a theoretical mechanism for the transient emergence of such drug tolerance. In this framework, we formulate an individual-based model and an integro-differential equation model of reversible phenotypic evolution in a cell population exposed to cytotoxic drugs. The outcomes of both models suggest that nongenetic instability, stress-induced adaptation, selection, and the interplay between these mechanisms can push an actively proliferating cell population to transition into a weakly proliferative and drug-tolerant state. Hence, the cell population experiences much less stress in the presence of the drugs and, in the long run, reacquires a proliferative phenotype, due to phenotypic fluctuations and selection pressure. These mechanisms can also reverse epigenetic drug tolerance following drug washout. Our study highlights how the transient appearance of the weakly proliferative and drug-tolerant cells is related to the use of high-dose therapy. Furthermore, we show how stem-like characteristics can act to stabilize the transient, weakly proliferative, and drug-tolerant subpopulation for a longer time window. Finally, using our models as in silico laboratories, we propose new testable hypotheses that could help uncover general principles underlying the emergence of cancer drug tolerance.
Collapse
Affiliation(s)
- Rebecca H Chisholm
- INRIA-Paris-Rocquencourt, MAMBA Team, Domaine de Voluceau, Le Chesnay Cedex, France. Sorbonne Universités, UPMC Univ Paris 06, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France. CNRS, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France.
| | - Tommaso Lorenzi
- INRIA-Paris-Rocquencourt, MAMBA Team, Domaine de Voluceau, Le Chesnay Cedex, France. Sorbonne Universités, UPMC Univ Paris 06, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France. CNRS, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France. CMLA, ENS Cachan, CNRS, PRES UniverSud, 61, Avenue du Président Wilson, Cachan Cedex, France
| | - Alexander Lorz
- INRIA-Paris-Rocquencourt, MAMBA Team, Domaine de Voluceau, Le Chesnay Cedex, France. Sorbonne Universités, UPMC Univ Paris 06, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France. CNRS, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France
| | - Annette K Larsen
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France. INSERM, UMR_S 938, Laboratory of Cancer Biology and Therapeutics, Paris, France
| | - Luís Neves de Almeida
- INRIA-Paris-Rocquencourt, MAMBA Team, Domaine de Voluceau, Le Chesnay Cedex, France. Sorbonne Universités, UPMC Univ Paris 06, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France. CNRS, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France
| | - Alexandre Escargueil
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France. INSERM, UMR_S 938, Laboratory of Cancer Biology and Therapeutics, Paris, France
| | - Jean Clairambault
- INRIA-Paris-Rocquencourt, MAMBA Team, Domaine de Voluceau, Le Chesnay Cedex, France. Sorbonne Universités, UPMC Univ Paris 06, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France. CNRS, UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France
| |
Collapse
|
183
|
Helmstetter C, Flossdorf M, Peine M, Kupz A, Zhu J, Hegazy AN, Duque-Correa MA, Zhang Q, Vainshtein Y, Radbruch A, Kaufmann SH, Paul WE, Höfer T, Löhning M. Individual T helper cells have a quantitative cytokine memory. Immunity 2015; 42:108-22. [PMID: 25607461 PMCID: PMC4562415 DOI: 10.1016/j.immuni.2014.12.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 11/14/2014] [Accepted: 12/19/2014] [Indexed: 12/24/2022]
Abstract
The probabilistic expression of cytokine genes in differentiated T helper (Th) cell populations remains ill defined. By single-cell analyses and mathematical modeling, we show that one stimulation featured stable cytokine nonproducers as well as stable producers with wide cell-to-cell variability in the magnitude of expression. Focusing on interferon-γ (IFN-γ) expression by Th1 cells, mathematical modeling predicted that this behavior reflected different cell-intrinsic capacities and not mere gene-expression noise. In vivo, Th1 cells sort purified by secreted IFN-γ amounts preserved a quantitative memory for both probability and magnitude of IFN-γ re-expression for at least 1 month. Mechanistically, this memory resulted from quantitatively distinct transcription of individual alleles and was controlled by stable expression differences of the Th1 cell lineage-specifying transcription factor T-bet. Functionally, Th1 cells with graded IFN-γ production competence differentially activated Salmonella-infected macrophages for bacterial killing. Thus, individual Th cells commit to produce distinct amounts of a given cytokine, thereby generating functional intrapopulation heterogeneity.
Collapse
Affiliation(s)
- Caroline Helmstetter
- Experimental Immunology, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, 10117 Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - Michael Flossdorf
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Bioquant Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Michael Peine
- Experimental Immunology, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, 10117 Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - Andreas Kupz
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Queensland Tropical Health Alliance Research Laboratory, James Cook University, Cairns Campus, Smithfield, QLD 4878, Australia
| | - Jinfang Zhu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Ahmed N Hegazy
- Experimental Immunology, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, 10117 Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany; Department of Gastroenterology, Hepatology and Endocrinology, Charité, 10117 Berlin, Germany
| | - Maria A Duque-Correa
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Qin Zhang
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Bioquant Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Yevhen Vainshtein
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Bioquant Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Andreas Radbruch
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - Stefan H Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - William E Paul
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Bioquant Center, University of Heidelberg, 69120 Heidelberg, Germany.
| | - Max Löhning
- Experimental Immunology, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, 10117 Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany.
| |
Collapse
|
184
|
Deconstructing transcriptional heterogeneity in pluripotent stem cells. Nature 2015; 516:56-61. [PMID: 25471879 PMCID: PMC4256722 DOI: 10.1038/nature13920] [Citation(s) in RCA: 270] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 10/07/2014] [Indexed: 01/15/2023]
Abstract
Pluripotent stem cells (PSCs) are capable of dynamic interconversion between distinct substates, but the regulatory circuits specifying these states and enabling transitions between them are not well understood. We set out to characterize transcriptional heterogeneity in PSCs by single-cell expression profiling under different chemical and genetic perturbations. Signaling factors and developmental regulators show highly variable expression, with expression states for some variable genes heritable through multiple cell divisions. Expression variability and population heterogeneity can be influenced by perturbation of signaling pathways and chromatin regulators. Strikingly, either removal of mature miRNAs or pharmacologic blockage of signaling pathways drives PSCs into a low-noise ground state characterized by a reconfigured pluripotency network, enhanced self-renewal, and a distinct chromatin state, an effect mediated by opposing miRNA families acting on the c-myc / Lin28 / let-7 axis. These data illuminate the nature of transcriptional heterogeneity in PSCs.
Collapse
|
185
|
Mackey MC, Santillán M, Tyran-Kamińska M, Zeron ES. The utility of simple mathematical models in understanding gene regulatory dynamics. In Silico Biol 2015; 12:23-53. [PMID: 25402755 PMCID: PMC4923710 DOI: 10.3233/isb-140463] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/22/2014] [Accepted: 10/23/2014] [Indexed: 11/17/2022]
Abstract
In this review, we survey work that has been carried out in the attempts of biomathematicians to understand the dynamic behaviour of simple bacterial operons starting with the initial work of the 1960's. We concentrate on the simplest of situations, discussing both repressible and inducible systems and then turning to concrete examples related to the biology of the lactose and tryptophan operons. We conclude with a brief discussion of the role of both extrinsic noise and so-called intrinsic noise in the form of translational and/or transcriptional bursting.
Collapse
Affiliation(s)
- Michael C. Mackey
- Departments of Physiology, Physics & Mathematics, McGill University, Montreal, Quebec, Canada
| | - Moisés Santillán
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Monterrey, Parque de Investigación e Innovación Tecnológica, Apodaca NL, México
| | | | - Eduardo S. Zeron
- Departamento de Matemáticas, Centro de Investigación y de Estudios Avanzados del IPN, Apartado Postal, México DF, México
| |
Collapse
|
186
|
Kempe H, Schwabe A, Crémazy F, Verschure PJ, Bruggeman FJ. The volumes and transcript counts of single cells reveal concentration homeostasis and capture biological noise. Mol Biol Cell 2014; 26:797-804. [PMID: 25518937 PMCID: PMC4325848 DOI: 10.1091/mbc.e14-08-1296] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
We present data on cell-to-cell variability (“‘noise”') of gene expression in human cells, obtained through a combination of single-molecule mRNA FISH and single-cell volume measurements. We find that noise in terms of mRNA numbers exceeds the noise in terms of mRNA concentration. This study provides an improved method to determine gene expression noise. Transcriptional stochasticity can be measured by counting the number of mRNA molecules per cell. Cell-to-cell variability is best captured in terms of concentration rather than molecule counts, because reaction rates depend on concentrations. We combined single-molecule mRNA counting with single-cell volume measurements to quantify the statistics of both transcript numbers and concentrations in human cells. We compared three cell clones that differ only in the genomic integration site of an identical constitutively expressed reporter gene. The transcript number per cell varied proportionally with cell volume in all three clones, indicating concentration homeostasis. We found that the cell-to-cell variability in the mRNA concentration is almost exclusively due to cell-to-cell variation in gene expression activity, whereas the cell-to-cell variation in mRNA number is larger, due to a significant contribution of cell volume variability. We concluded that the precise relationship between transcript number and cell volume sets the biological stochasticity of living cells. This study highlights the importance of the quantitative measurement of transcript concentrations in studies of cell-to-cell variability in biology.
Collapse
Affiliation(s)
- Hermannus Kempe
- Synthetic Systems Biology and Nuclear Organization Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Anne Schwabe
- Systems Bioinformatics, Amsterdam Institute for Molecules, Medicines and Systems, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Frédéric Crémazy
- Synthetic Systems Biology and Nuclear Organization Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Pernette J Verschure
- Synthetic Systems Biology and Nuclear Organization Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Frank J Bruggeman
- Systems Bioinformatics, Amsterdam Institute for Molecules, Medicines and Systems, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
187
|
Tarabichi M, Antoniou A, Saiselet M, Pita JM, Andry G, Dumont JE, Detours V, Maenhaut C. Systems biology of cancer: entropy, disorder, and selection-driven evolution to independence, invasion and "swarm intelligence". Cancer Metastasis Rev 2014; 32:403-21. [PMID: 23615877 PMCID: PMC3843370 DOI: 10.1007/s10555-013-9431-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Our knowledge of the biology of solid cancer has greatly progressed during the last few years, and many excellent reviews dealing with the various aspects of this biology have appeared. In the present review, we attempt to bring together these subjects in a general systems biology narrative. It starts from the roles of what we term entropy of signaling and noise in the initial oncogenic events, to the first major transition of tumorigenesis: the independence of the tumor cell and the switch in its physiology, i.e., from subservience to the organism to its own independent Darwinian evolution. The development after independence involves a constant dynamic reprogramming of the cells and the emergence of a sort of collective intelligence leading to invasion and metastasis and seldom to the ultimate acquisition of immortality through inter-individual infection. At each step, the probability of success is minimal to infinitesimal, but the number of cells possibly involved and the time scale account for the relatively high occurrence of tumorigenesis and metastasis in multicellular organisms.
Collapse
Affiliation(s)
| | | | | | - J. M. Pita
- IRIBHM, Brussels, Belgium
- UIPM, Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOFG) and CEDOC, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - G. Andry
- J. Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | | | | | - C. Maenhaut
- IRIBHM, Brussels, Belgium
- WELBIO, Wallonia, Belgium
| |
Collapse
|
188
|
Ståhlberg A, Kubista M. The workflow of single-cell expression profiling using quantitative real-time PCR. Expert Rev Mol Diagn 2014; 14:323-31. [PMID: 24649819 PMCID: PMC4819576 DOI: 10.1586/14737159.2014.901154] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Biological material is heterogeneous and when exposed to stimuli the various cells present respond differently. Much of the complexity can be eliminated by disintegrating the sample, studying the cells one by one. Single-cell profiling reveals responses that go unnoticed when classical samples are studied. New cell types and cell subtypes may be found and relevant pathways and expression networks can be identified. The most powerful technique for single-cell expression profiling is currently quantitative reverse transcription real-time PCR (RT-qPCR). A robust RT-qPCR workflow for highly sensitive and specific measurements in high-throughput and a reasonable degree of multiplexing has been developed for targeting mRNAs, but also microRNAs, non-coding RNAs and most recently also proteins. We review the current state of the art of single-cell expression profiling and present also the improvements and developments expected in the next 5 years.
Collapse
Affiliation(s)
- Anders Ståhlberg
- 1Department of Pathology, Sahlgrenska Cancer Center, University of Gothenburg, Box 425, 40530 Gothenburg, Sweden
| | | |
Collapse
|
189
|
Singer ZS, Yong J, Tischler J, Hackett JA, Altinok A, Surani MA, Cai L, Elowitz MB. Dynamic heterogeneity and DNA methylation in embryonic stem cells. Mol Cell 2014; 55:319-31. [PMID: 25038413 PMCID: PMC4104113 DOI: 10.1016/j.molcel.2014.06.029] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 04/04/2014] [Accepted: 06/18/2014] [Indexed: 01/16/2023]
Abstract
Cell populations can be strikingly heterogeneous, composed of multiple cellular states, each exhibiting stochastic noise in its gene expression. A major challenge is to disentangle these two types of variability and to understand the dynamic processes and mechanisms that control them. Embryonic stem cells (ESCs) provide an ideal model system to address this issue because they exhibit heterogeneous and dynamic expression of functionally important regulatory factors. We analyzed gene expression in individual ESCs using single-molecule RNA-FISH and quantitative time-lapse movies. These data discriminated stochastic switching between two coherent (correlated) gene expression states and burst-like transcriptional noise. We further showed that the “2i” signaling pathway inhibitors modulate both types of variation. Finally, we found that DNA methylation plays a key role in maintaining these metastable states. Together, these results show how ESC gene expression states and dynamics arise from a combination of intrinsic noise, coherent cellular states, and epigenetic regulation. smFISH in ESCs reveals two transcriptional states and highly stochastic expression Live-cell expression dynamics reveal the in situ transition rates between states DNA methylation regulates state-switching dynamics “2i” signaling inhibitors impact both gene expression noise and state transitions
Collapse
Affiliation(s)
- Zakary S Singer
- Computation and Neural Systems, California Institute of Technology, Pasadena, CA 91125, USA
| | - John Yong
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Julia Tischler
- The Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Jamie A Hackett
- The Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Alphan Altinok
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA; Biological Network Modeling Center, California Institute of Technology, Pasadena, CA 91125, USA
| | - M Azim Surani
- The Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Long Cai
- Program in Biochemistry and Molecular Biophysics and Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael B Elowitz
- Howard Hughes Medical Institute and Division of Biology and Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
190
|
Bertaux F, Stoma S, Drasdo D, Batt G. Modeling dynamics of cell-to-cell variability in TRAIL-induced apoptosis explains fractional killing and predicts reversible resistance. PLoS Comput Biol 2014; 10:e1003893. [PMID: 25340343 PMCID: PMC4207462 DOI: 10.1371/journal.pcbi.1003893] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/04/2014] [Indexed: 12/22/2022] Open
Abstract
Isogenic cells sensing identical external signals can take markedly different decisions. Such decisions often correlate with pre-existing cell-to-cell differences in protein levels. When not neglected in signal transduction models, these differences are accounted for in a static manner, by assuming randomly distributed initial protein levels. However, this approach ignores the a priori non-trivial interplay between signal transduction and the source of this cell-to-cell variability: temporal fluctuations of protein levels in individual cells, driven by noisy synthesis and degradation. Thus, modeling protein fluctuations, rather than their consequences on the initial population heterogeneity, would set the quantitative analysis of signal transduction on firmer grounds. Adopting this dynamical view on cell-to-cell differences amounts to recast extrinsic variability into intrinsic noise. Here, we propose a generic approach to merge, in a systematic and principled manner, signal transduction models with stochastic protein turnover models. When applied to an established kinetic model of TRAIL-induced apoptosis, our approach markedly increased model prediction capabilities. One obtains a mechanistic explanation of yet-unexplained observations on fractional killing and non-trivial robust predictions of the temporal evolution of cell resistance to TRAIL in HeLa cells. Our results provide an alternative explanation to survival via induction of survival pathways since no TRAIL-induced regulations are needed and suggest that short-lived anti-apoptotic protein Mcl1 exhibit large and rare fluctuations. More generally, our results highlight the importance of accounting for stochastic protein turnover to quantitatively understand signal transduction over extended durations, and imply that fluctuations of short-lived proteins deserve particular attention.
Collapse
Affiliation(s)
| | | | - Dirk Drasdo
- INRIA Paris-Rocquencourt, Le Chesnay, France
- Laboratoire Jacques-Louis Lions (LJLL), University of Paris 6 (UPMC) - CNRS (UMR7598), Paris, France
| | - Gregory Batt
- INRIA Paris-Rocquencourt, Le Chesnay, France
- * E-mail:
| |
Collapse
|
191
|
Davis DM, Purvis JE. Computational analysis of signaling patterns in single cells. Semin Cell Dev Biol 2014; 37:35-43. [PMID: 25263011 DOI: 10.1016/j.semcdb.2014.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/11/2014] [Accepted: 09/13/2014] [Indexed: 01/19/2023]
Abstract
Signaling proteins are flexible in both form and function. They can bind to multiple molecular partners and integrate diverse types of cellular information. When imaged by time-lapse microscopy, many signaling proteins show complex patterns of activity or localization that vary from cell to cell. This heterogeneity is so prevalent that it has spurred the development of new computational strategies to analyze single-cell signaling patterns. A collective observation from these analyses is that cells appear less heterogeneous when their responses are normalized to, or synchronized with, other single-cell measurements. In many cases, these transformed signaling patterns show distinct dynamical trends that correspond with predictable phenotypic outcomes. When signaling mechanisms are unclear, computational models can suggest putative molecular interactions that are experimentally testable. Thus, computational analysis of single-cell signaling has not only provided new ways to quantify the responses of individual cells, but has helped resolve longstanding questions surrounding many well-studied human signaling proteins including NF-κB, p53, ERK1/2, and CDK2. A number of specific challenges lie ahead for single-cell analysis such as quantifying the contribution of non-cell autonomous signaling as well as the characterization of protein signaling dynamics in vivo.
Collapse
Affiliation(s)
- Denise M Davis
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599-7264, United States
| | - Jeremy E Purvis
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599-7264, United States.
| |
Collapse
|
192
|
Han L, Zi X, Garmire LX, Wu Y, Weissman SM, Pan X, Fan R. Co-detection and sequencing of genes and transcripts from the same single cells facilitated by a microfluidics platform. Sci Rep 2014; 4:6485. [PMID: 25255798 PMCID: PMC4175731 DOI: 10.1038/srep06485] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 09/05/2014] [Indexed: 01/12/2023] Open
Abstract
Despite the recent advance of single-cell gene expression analyses, co-measurement of both genomic and transcriptional signatures at the single-cell level has not been realized. However such analysis is necessary in order to accurately delineate how genetic information is transcribed, expressed, and regulated to give rise to an enormously diverse range of cell phenotypes. Here we report on a microfluidics-facilitated approach that allows for controlled separation of cytoplasmic and nuclear contents of a single cell followed by on-chip amplification of genomic DNA and cytoplasmic mRNA. When coupled with off-chip polymerase chain reaction, gel electrophoresis and Sanger sequencing, a panel of genes and transcripts from the same single cell can be co-detected and sequenced. This platform is potentially an enabling tool to permit multiple genomic measurements performed on the same single cells and opens new opportunities to tackle a range of fundamental biology questions including non-genetic cell-to-cell variability, epigenetic regulation, and stem cell fate control. It also helps address clinical challenges such as diagnosing intra-tumor heterogeneity and dissecting complex cellular immune responses.
Collapse
Affiliation(s)
- Lin Han
- 1] Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA [2]
| | - Xiaoyuan Zi
- 1] Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA [2] Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA [3] Department of Cell Biology, Second Military Medical University, Shanghai 200433, China [4]
| | - Lana X Garmire
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI 96813 USA
| | - Yu Wu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Sherman M Weissman
- 1] Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA [2] Yale Comprehensive Cancer Center, New Haven, CT 06520, USA
| | - Xinghua Pan
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Rong Fan
- 1] Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA [2] Yale Comprehensive Cancer Center, New Haven, CT 06520, USA
| |
Collapse
|
193
|
Abstract
Any organism is embedded in an environment that changes over time. The timescale for and statistics of environmental change, the precision with which the organism can detect its environment, and the costs and benefits of particular protein expression levels all will affect the suitability of different strategies--such as constitutive expression or graded response--for regulating protein levels in response to environmental inputs. We propose a general framework-here specifically applied to the enzymatic regulation of metabolism in response to changing concentrations of a basic nutrient-to predict the optimal regulatory strategy given the statistics of fluctuations in the environment and measurement apparatus, respectively, and the costs associated with enzyme production. We use this framework to address three fundamental questions: (i) when a cell should prefer thresholding to a graded response; (ii) when there is a fitness advantage to implementing a Bayesian decision rule; and (iii) when retaining memory of the past provides a selective advantage. We specifically find that: (i) relative convexity of enzyme expression cost and benefit influences the fitness of thresholding or graded responses; (ii) intermediate levels of measurement uncertainty call for a sophisticated Bayesian decision rule; and (iii) in dynamic contexts, intermediate levels of uncertainty call for retaining memory of the past. Statistical properties of the environment, such as variability and correlation times, set optimal biochemical parameters, such as thresholds and decay rates in signaling pathways. Our framework provides a theoretical basis for interpreting molecular signal processing algorithms and a classification scheme that organizes known regulatory strategies and may help conceptualize heretofore unknown ones.
Collapse
|
194
|
Kotelnikova E, Bernardo-Faura M, Silberberg G, Kiani NA, Messinis D, Melas IN, Artigas L, Schwartz E, Mazo I, Masso M, Alexopoulos LG, Mas JM, Olsson T, Tegner J, Martin R, Zamora A, Paul F, Saez-Rodriguez J, Villoslada P. Signaling networks in MS: a systems-based approach to developing new pharmacological therapies. Mult Scler 2014; 21:138-46. [PMID: 25112814 DOI: 10.1177/1352458514543339] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pathogenesis of multiple sclerosis (MS) involves alterations to multiple pathways and processes, which represent a significant challenge for developing more-effective therapies. Systems biology approaches that study pathway dysregulation should offer benefits by integrating molecular networks and dynamic models with current biological knowledge for understanding disease heterogeneity and response to therapy. In MS, abnormalities have been identified in several cytokine-signaling pathways, as well as those of other immune receptors. Among the downstream molecules implicated are Jak/Stat, NF-Kb, ERK1/3, p38 or Jun/Fos. Together, these data suggest that MS is likely to be associated with abnormalities in apoptosis/cell death, microglia activation, blood-brain barrier functioning, immune responses, cytokine production, and/or oxidative stress, although which pathways contribute to the cascade of damage and can be modulated remains an open question. While current MS drugs target some of these pathways, others remain untouched. Here, we propose a pragmatic systems analysis approach that involves the large-scale extraction of processes and pathways relevant to MS. These data serve as a scaffold on which computational modeling can be performed to identify disease subgroups based on the contribution of different processes. Such an analysis, targeting these relevant MS-signaling pathways, offers the opportunity to accelerate the development of novel individual or combination therapies.
Collapse
Affiliation(s)
- Ekaterina Kotelnikova
- Institute Biomedical Research August Pi Sunyer (IDIBAPS) - Hospital Clinic of Barcelona, Spain/Personal Biomedicine ZAO, and A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Russia
| | | | - Gilad Silberberg
- Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Sweden
| | - Narsis A Kiani
- Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Sweden
| | | | - Ioannis N Melas
- European Molecular Biology Laboratory, European Bioinformatics Institute, UK/ProtATonce Ltd, Greece/National Technical University of Athens, Greece
| | | | | | | | | | | | | | | | - Jesper Tegner
- Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Sweden
| | | | | | - Friedemann Paul
- NeuroCure Clinical Research Center and Department of Neurology, Charité University Medicine Berlin, Germany
| | | | - Pablo Villoslada
- Institute Biomedical Research August Pi Sunyer (IDIBAPS) - Hospital Clinic of Barcelona, Spain/Personal Biomedicine ZAO, and A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Russia
| |
Collapse
|
195
|
Bordi I, Ricigliano VAG, Umeton R, Ristori G, Grassi F, Crisanti A, Sutera A, Salvetti M. Noise in multiple sclerosis: unwanted and necessary. Ann Clin Transl Neurol 2014; 1:502-11. [PMID: 25356421 PMCID: PMC4184780 DOI: 10.1002/acn3.72] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/12/2014] [Accepted: 05/17/2014] [Indexed: 12/25/2022] Open
Abstract
As our knowledge about the etiology of multiple sclerosis (MS) increases, deterministic paradigms appear insufficient to describe the pathogenesis of the disease, and the impression is that stochastic phenomena (i.e. random events not necessarily resulting in disease in all individuals) may contribute to the development of MS. However, sources and mechanisms of stochastic behavior have not been investigated and there is no proposed framework to incorporate nondeterministic processes into disease biology. In this report, we will first describe analogies between physics of nonlinear systems and cell biology, showing how small-scale random perturbations can impact on large-scale phenomena, including cell function. We will then review growing and solid evidence showing that stochastic gene expression (or gene expression “noise”) can be a driver of phenotypic variation. Moreover, we will describe new methods that open unprecedented opportunities for the study of such phenomena in patients and the impact of this information on our understanding of MS course and therapy.
Collapse
Affiliation(s)
- Isabella Bordi
- Department of Physics, Sapienza University of Rome Rome, Italy
| | - Vito A G Ricigliano
- Neurology and Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome Rome, Italy ; Neuroimmunology Unit, Fondazione Santa Lucia, (I.R.C.C.S.) Rome, Italy
| | - Renato Umeton
- Neurology and Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome Rome, Italy
| | - Giovanni Ristori
- Neurology and Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome Rome, Italy
| | - Francesca Grassi
- Department of Physiology and Pharmacology, Sapienza University of Rome Rome, Italy
| | - Andrea Crisanti
- Department of Physics, Sapienza University of Rome Rome, Italy
| | - Alfonso Sutera
- Department of Physics, Sapienza University of Rome Rome, Italy
| | - Marco Salvetti
- Neurology and Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome Rome, Italy
| |
Collapse
|
196
|
Torres-Padilla ME, Chambers I. Transcription factor heterogeneity in pluripotent stem cells: a stochastic advantage. Development 2014; 141:2173-81. [PMID: 24866112 DOI: 10.1242/dev.102624] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
When pluripotent cells are exposed to a uniform culture environment they routinely display heterogeneous gene expression. Aspects of this heterogeneity, such as Nanog expression, are linked to differences in the propensity of individual cells to either self-renew or commit towards differentiation. Recent findings have provided new insight into the underlying causes of this heterogeneity, which we summarise here using Nanog, a key regulator of pluripotency, as a model gene. We discuss the role of transcription factor heterogeneity in facilitating the intrinsically dynamic and stochastic nature of the pluripotency network, which in turn provides a potential benefit to a population of cells that needs to balance cell fate decisions.
Collapse
Affiliation(s)
- Maria-Elena Torres-Padilla
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM U964, Université de Strasbourg, Cité Universitaire de Strasbourg, Illkirch F-67404, France
| | - Ian Chambers
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
197
|
Strovas TJ, Rosenberg AB, Kuypers BE, Muscat RA, Seelig G. MicroRNA-based single-gene circuits buffer protein synthesis rates against perturbations. ACS Synth Biol 2014; 3:324-31. [PMID: 24847681 DOI: 10.1021/sb4001867] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Achieving precise control of mammalian transgene expression has remained a long-standing, and increasingly urgent, challenge in biomedical science. Despite much work, single-cell methods have consistently revealed that mammalian gene expression levels remain susceptible to fluctuations (noise) and external perturbations. Here, we show that precise control of protein synthesis can be realized using a single-gene microRNA (miRNA)-based feed-forward loop (sgFFL). This minimal autoregulatory gene circuit consists of an intronic miRNA that targets its own transcript. In response to a step-like increase in transcription rate, the network generated a transient protein expression pulse before returning to a lower steady state level, thus exhibiting adaptation. Critically, the steady state protein levels were independent of the size of the stimulus, demonstrating that this simple network architecture effectively buffered protein production against changes in transcription. The single-gene network architecture was also effective in buffering against transcriptional noise, leading to reduced cell-to-cell variability in protein synthesis. Noise was up to 5-fold lower for a sgFFL than for an unregulated control gene with equal mean protein levels. The noise buffering capability varied predictably with the strength of the miRNA-target interaction. Together, these results suggest that the sgFFL single-gene motif provides a general and broadly applicable platform for robust gene expression in synthetic and natural gene circuits.
Collapse
Affiliation(s)
- Timothy J Strovas
- Department of Electrical Engineering and ‡Department of Computer Science & Engineering, University of Washington , Seattle, Washington 98195-5852, United States
| | | | | | | | | |
Collapse
|
198
|
Charlebois DA, Balázsi G, Kærn M. Coherent feedforward transcriptional regulatory motifs enhance drug resistance. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2014; 89:052708. [PMID: 25353830 PMCID: PMC5749921 DOI: 10.1103/physreve.89.052708] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Indexed: 05/25/2023]
Abstract
Fluctuations in gene expression give identical cells access to a spectrum of phenotypes that can serve as a transient, nongenetic basis for natural selection by temporarily increasing drug resistance. In this study, we demonstrate using mathematical modeling and simulation that certain gene regulatory network motifs, specifically coherent feedforward loop motifs, can facilitate the development of nongenetic resistance by increasing cell-to-cell variability and the time scale at which beneficial phenotypic states can be maintained. Our results highlight how regulatory network motifs enabling transient, nongenetic inheritance play an important role in defining reproductive fitness in adverse environments and provide a selective advantage subject to evolutionary pressure.
Collapse
Affiliation(s)
- Daniel A Charlebois
- Department of Physics, University of Ottawa, 150 Louis Pasteur, Ottawa, Ontario, Canada K1N 6N5 and Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, Canada K1H 8M5
| | - Gábor Balázsi
- Department of Systems Biology-Unit 950, University of Texas MD Anderson Cancer Center, 7435 Fannin Street, Houston, Texas 77054, USA
| | - Mads Kærn
- Department of Physics, University of Ottawa, 150 Louis Pasteur, Ottawa, Ontario, Canada K1N 6N5 and Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, Canada K1H 8M5 and Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
199
|
Abstract
The tight regulation of splicing networks is critical to development. To maintain robust splicing patterns, many splicing factors autoregulate their expression through alternative splicing-coupled nonsense-mediated decay (AS-NMD). Here, Jangi et al. globally characterize the function of the splicing regulator Rbfox2 in mouse embryonic stem cells. They show that Rbfox2 cross-regulates AS-NMD events within RNA-binding proteins to alter their expression. This study positions Rbfox2 at a critical node in a broader splicing regulatory network, with roles in both normal development and disease. The tight regulation of splicing networks is critical for organismal development. To maintain robust splicing patterns, many splicing factors autoregulate their expression through alternative splicing-coupled nonsense-mediated decay (AS-NMD). However, as negative autoregulation results in a self-limiting window of splicing factor expression, it is unknown how variations in steady-state protein levels can arise in different physiological contexts. Here, we demonstrate that Rbfox2 cross-regulates AS-NMD events within RNA-binding proteins to alter their expression. Using individual nucleotide-resolution cross-linking immunoprecipitation coupled to high-throughput sequencing (iCLIP) and mRNA sequencing, we identified >200 AS-NMD splicing events that are bound by Rbfox2 in mouse embryonic stem cells. These “silent” events are characterized by minimal apparent splicing changes but appreciable changes in gene expression upon Rbfox2 knockdown due to degradation of the NMD-inducing isoform. Nearly 70 of these AS-NMD events fall within genes encoding RNA-binding proteins, many of which are autoregulated. As with the coding splicing events that we found to be regulated by Rbfox2, silent splicing events are evolutionarily conserved and frequently contain the Rbfox2 consensus UGCAUG. Our findings uncover an unexpectedly broad and multilayer regulatory network controlled by Rbfox2 and offer an explanation for how autoregulatory splicing networks are tuned.
Collapse
|
200
|
Chai J, Song Q. Multiple-protein detections of single-cells reveal cell-cell heterogeneity in human cells. IEEE Trans Biomed Eng 2014; 62:30-8. [PMID: 24710818 DOI: 10.1109/tbme.2014.2315437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cell population represents an intrinsically heterogeneous and stochastic system, in which individual cells often behave very differently in molecular contents, functions and even genotypes from the population average in response to uniform physiological stimuli. The traditional bulk cellular analysis often overlooks cellular heterogeneity and does not provide information on cell-cell variations. Single-cell measurements can reveal information obscured in population averages, and enable us to determine distributions rather than averaged properties within a cell population. The level of complexity, with numerous variables acting at the same time, requires multiparametric and dynamic investigation of a large number of single cells. Multiplexed study can provide quantitative correlations or inter-relationships among multiple cellular components and molecular markers within a protein network or family in biological processes. In this paper, we applied multiple fluorophore-conjugated primary antibodies to detect multiple proteins expressed on the same singe cells from a clonal population. To reveal cell-cell heterogeneity, we quantified the histograms of six proteins within a cell population as functions of TNF-α stimulation time. Then, we quantified noise and noise strength of these protein histograms as functions of TNF-α stimulation time. Thirdly, we quantified correlation coefficients of multiple proteins expressed on same single-cells as functions of TNF-α stimulation time. Above parameters demonstrated nonlinear relationships with TNF-α stimulation. Quantification of above parameters on independent cell subpopulations further reveals the cell-cell heterogeneity when exposed to identical environmental conditions. Such cellular heterogeneity will be useful to characterize the disease progression and disease diagnoses.
Collapse
|