Frequency modulation of ERK activation dynamics rewires cell fate

Transient versus sustained ERK MAP kinase (MAPK) activation dynamics induce proliferation versus differentiation in response to epidermal (EGF) or nerve (NGF) growth factors in PC‐12 cells. Duration of ERK activation has therefore been proposed to specify cell fate decisions. Using a biosensor to measure ERK activation dynamics in single living cells reveals that sustained EGF/NGF application leads to a heterogeneous mix of transient and sustained ERK activation dynamics in distinct cells of the population, different than the population average. EGF biases toward transient, while NGF biases toward sustained ERK activation responses. In contrast, pulsed growth factor application can repeatedly and homogeneously trigger ERK activity transients across the cell population. These datasets enable mathematical modeling to reveal salient features inherent to the MAPK network. Ultimately, this predicts pulsed growth factor stimulation regimes that can bypass the typical feedback activation to rewire the system toward cell differentiation irrespective of growth factor identity.

[1]  K. Aoki,et al.  Fluorescence resonance energy transfer based quantitative analysis of feedforward and feedback loops in epidermal growth factor receptor signaling and the sensitivity to molecular targeting drugs , 2014, The FEBS journal.

[2]  Laura E. Edwards,et al.  Supplementary Figure Legends , 2022 .

[3]  M. Elowitz,et al.  Functional Roles of Pulsing in Genetic Circuits , 2013, Science.

[4]  Kazuhiro Aoki,et al.  Stochastic ERK activation induced by noise and cell-to-cell propagation regulates cell density-dependent proliferation. , 2013, Molecular cell.

[5]  Jacco van Rheenen,et al.  A Versatile Toolkit to Produce Sensitive FRET Biosensors to Visualize Signaling in Time and Space , 2013, Science Signaling.

[6]  G. Lahav,et al.  Encoding and Decoding Cellular Information through Signaling Dynamics , 2013, Cell.

[7]  S. Cagnol,et al.  Oncogenic KRAS and BRAF activation of the MEK/ERK signaling pathway promotes expression of dual-specificity phosphatase 4 (DUSP4/MKP2) resulting in nuclear ERK1/2 inhibition , 2013, Oncogene.

[8]  John G. Albeck,et al.  Frequency-modulated pulses of ERK activity transmit quantitative proliferation signals. , 2013, Molecular cell.

[9]  Boris N. Kholodenko,et al.  Crosstalk and Signaling Switches in Mitogen-Activated Protein Kinase Cascades , 2012, Front. Physiol..

[10]  Luke P. Lee,et al.  Quantitative and dynamic assay of single cell chemotaxis. , 2012, Integrative biology : quantitative biosciences from nano to macro.

[11]  Tobias Meyer,et al.  Supplemental Experimental Procedures A Two-Dimensional ERK-AKT Signaling Code for an NGF-Triggered Cell-Fate Decision , 2011 .

[12]  Marc R. Birtwistle,et al.  Linear Approaches to Intramolecular Förster Resonance Energy Transfer Probe Measurements for Quantitative Modeling , 2011, PloS one.

[13]  Jin Zhang,et al.  Spatiotemporally Regulated Protein Kinase A Activity Is a Critical Regulator of Growth Factor-Stimulated Extracellular Signal-Regulated Kinase Signaling in PC12 Cells , 2011, Molecular and Cellular Biology.

[14]  Nils Blüthgen,et al.  Strong negative feedback from Erk to Raf confers robustness to MAPK signalling , 2011, Molecular systems biology.

[15]  Anne E Carpenter,et al.  Improved structure, function and compatibility for CellProfiler: modular high-throughput image analysis software , 2011, Bioinform..

[16]  W. Kolch,et al.  Raf family kinases: old dogs have learned new tricks. , 2011, Genes & cancer.

[17]  Yosef Yarden,et al.  Feedback regulation of EGFR signalling: decision making by early and delayed loops , 2011, Nature Reviews Molecular Cell Biology.

[18]  B. Snijder,et al.  Origins of regulated cell-to-cell variability , 2011, Nature Reviews Molecular Cell Biology.

[19]  G. Pagès,et al.  Post‐transcriptional regulation of the DUSP6/MKP‐3 phosphatase by MEK/ERK signaling and hypoxia , 2011, Journal of cellular physiology.

[20]  Boris N. Kholodenko,et al.  Signalling ballet in space and time , 2010, Nature Reviews Molecular Cell Biology.

[21]  Boris N. Kholodenko,et al.  Ligand-Specific c-Fos Expression Emerges from the Spatiotemporal Control of ErbB Network Dynamics , 2010, Cell.

[22]  A. Sorkin,et al.  Quantitative Analysis of Endocytosis and Turnover of Epidermal Growth Factor (EGF) and EGF Receptor , 2010, Current protocols in cell biology.

[23]  Uri Alon,et al.  Dynamics and variability of ERK2 response to EGF in individual living cells. , 2009, Molecular cell.

[24]  Walter Kolch,et al.  Cell fate decisions are specified by the dynamic ERK interactome , 2009, Nature Cell Biology.

[25]  K. Patterson,et al.  Dual-specificity phosphatases: critical regulators with diverse cellular targets. , 2009, The Biochemical journal.

[26]  Kwang-Hyun Cho,et al.  Positive- and negative-feedback regulations coordinate the dynamic behavior of the Ras-Raf-MEK-ERK signal transduction pathway , 2009, Journal of Cell Science.

[27]  K. Svoboda,et al.  A genetically encoded fluorescent sensor of ERK activity , 2008, Proceedings of the National Academy of Sciences.

[28]  Julio Saez-Rodriguez,et al.  Automatic decomposition of kinetic models of signaling networks minimizing the retroactivity among modules , 2008, ECCB.

[29]  Eduardo Sontag,et al.  Modular cell biology: retroactivity and insulation , 2008, Molecular systems biology.

[30]  P. Bastiaens,et al.  Growth factor-induced MAPK network topology shapes Erk response determining PC-12 cell fate , 2007, Nature Cell Biology.

[31]  Xuejun Jiang,et al.  Differential regulation of EGF receptor internalization and degradation by multiubiquitination within the kinase domain. , 2006, Molecular cell.

[32]  B. Kholodenko Cell-signalling dynamics in time and space , 2006, Nature Reviews Molecular Cell Biology.

[33]  Shinya Kuroda,et al.  Prediction and validation of the distinct dynamics of transient and sustained ERK activation , 2005, Nature Cell Biology.

[34]  E D Gilles,et al.  Reduction of mathematical models of signal transduction networks: simulation-based approach applied to EGF receptor signalling. , 2004, Systems biology.

[35]  Rey-Huei Chen,et al.  Molecular interpretation of ERK signal duration by immediate early gene products , 2002, Nature Cell Biology.

[36]  B. Kholodenko,et al.  Negative feedback and ultrasensitivity can bring about oscillations in the mitogen-activated protein kinase cascades. , 2000, European journal of biochemistry.

[37]  J E Ferrell,et al.  The biochemical basis of an all-or-none cell fate switch in Xenopus oocytes. , 1998, Science.

[38]  C. Marshall,et al.  Specificity of receptor tyrosine kinase signaling: Transient versus sustained extracellular signal-regulated kinase activation , 1995, Cell.

[39]  James E. Ferrell,et al.  A positive-feedback-based bistable ‘memory module’ that governs a cell fate decision , 2007, Nature.