1
|
Li Y, Deng D, Höfer CT, Kim J, Do Heo W, Xu Q, Liu X, Zi Z. Liebig's law of the minimum in the TGF-β/SMAD pathway. PLoS Comput Biol 2024; 20:e1012072. [PMID: 38753874 PMCID: PMC11135686 DOI: 10.1371/journal.pcbi.1012072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/29/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
Cells use signaling pathways to sense and respond to their environments. The transforming growth factor-β (TGF-β) pathway produces context-specific responses. Here, we combined modeling and experimental analysis to study the dependence of the output of the TGF-β pathway on the abundance of signaling molecules in the pathway. We showed that the TGF-β pathway processes the variation of TGF-β receptor abundance using Liebig's law of the minimum, meaning that the output-modifying factor is the signaling protein that is most limited, to determine signaling responses across cell types and in single cells. We found that the abundance of either the type I (TGFBR1) or type II (TGFBR2) TGF-β receptor determined the responses of cancer cell lines, such that the receptor with relatively low abundance dictates the response. Furthermore, nuclear SMAD2 signaling correlated with the abundance of TGF-β receptor in single cells depending on the relative expression levels of TGFBR1 and TGFBR2. A similar control principle could govern the heterogeneity of signaling responses in other signaling pathways.
Collapse
Affiliation(s)
- Yuchao Li
- Max Planck Institute for Molecular Genetics, Otto Warburg Laboratory, Berlin, Germany
| | - Difan Deng
- German Federal Institute for Risk Assessment, Department of Experimental Toxicology and ZEBET, Berlin, Germany
| | - Chris Tina Höfer
- German Federal Institute for Risk Assessment, Department of Experimental Toxicology and ZEBET, Berlin, Germany
| | - Jihye Kim
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Quanbin Xu
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Xuedong Liu
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Zhike Zi
- Max Planck Institute for Molecular Genetics, Otto Warburg Laboratory, Berlin, Germany
- German Federal Institute for Risk Assessment, Department of Experimental Toxicology and ZEBET, Berlin, Germany
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
2
|
Ghomlaghi M, Theocharous M, Hoang N, Shin SY, von Kriegsheim A, O’ Neill E, Zhang T, Nguyen LK. Integrative modeling and analysis of signaling crosstalk reveal molecular switches coordinating Yes-associated protein transcriptional activities. iScience 2024; 27:109031. [PMID: 38380257 PMCID: PMC10877689 DOI: 10.1016/j.isci.2024.109031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 12/07/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
The transcriptional co-activator YAP forms complexes with distinct transcription factors, controlling cell fate decisions, such as proliferation and apoptosis. However, the mechanisms underlying its context-dependent function are poorly defined. This study explores the interplay between the TGF-β and Hippo pathways and their influence on YAP's association with specific transcription factors. By integrating iterative mathematical modeling with experimental validation, we uncover molecular switches, predominantly controlled by RASSF1A and ITCH, which dictate the formation of YAP-SMAD (proliferative) and YAP-p73 (apoptotic) complexes. Our results show that RASSF1A enhances the formation of apoptotic complexes, whereas ITCH promotes the formation of proliferative complexes. Notably, higher levels of ITCH transform YAP-SMAD activity from a transient to a sustained state, impacting cellular behaviors. Extending these findings to various breast cancer cell lines highlights the role of cellular context in YAP regulation. Our study provides new insights into the mechanisms of YAP transcriptional activities and their therapeutic implications.
Collapse
Affiliation(s)
- Milad Ghomlaghi
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Mandy Theocharous
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Nhan Hoang
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Sung-Young Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alex von Kriegsheim
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Eric O’ Neill
- CRUK/MRC Institute for Radiation Oncology and Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Tao Zhang
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Lan K. Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
3
|
Richardson L, Wilcockson SG, Guglielmi L, Hill CS. Context-dependent TGFβ family signalling in cell fate regulation. Nat Rev Mol Cell Biol 2023; 24:876-894. [PMID: 37596501 DOI: 10.1038/s41580-023-00638-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/20/2023]
Abstract
The transforming growth factor-β (TGFβ) family are a large group of evolutionarily conserved cytokines whose signalling modulates cell fate decision-making across varying cellular contexts at different stages of life. Here we discuss new findings in early embryos that reveal how, in contrast to our original understanding of morphogen interpretation, robust cell fate specification can originate from a noisy combination of signalling inputs and a broad range of signalling levels. We compare this evidence with novel findings on the roles of TGFβ family signalling in tissue maintenance and homeostasis during juvenile and adult life, spanning the skeletal, haemopoietic and immune systems. From these comparisons, it emerges that in contrast to robust developing systems, relatively small perturbations in TGFβ family signalling have detrimental effects at later stages in life, leading to aberrant cell fate specification and disease, for example in cancer or congenital disorders. Finally, we highlight novel strategies to target and amend dysfunction in signalling and discuss how gleaning knowledge from different fields of biology can help in the development of therapeutics for aberrant TGFβ family signalling in disease.
Collapse
Affiliation(s)
- Louise Richardson
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
| | - Scott G Wilcockson
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
| | - Luca Guglielmi
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
4
|
Sofroniou MM, Lemmon CA. Differential regulation of fibronectin expression and fibrillogenesis by autocrine TGF-β1 signaling in malignant and benign mammary epithelial cells. Int J Biochem Cell Biol 2023; 165:106478. [PMID: 37866655 PMCID: PMC10775780 DOI: 10.1016/j.biocel.2023.106478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Remodeling of the extracellular matrix (ECM) is a key hallmark of cancer progression. A critical component of ECM remodeling is the assembly of the glycoprotein fibronectin (FN) into insoluble fibrils, which provide a scaffold for invading vascular endothelial cells and escaping cancer cells, as well as a framework for collagen deposition and oncogenic cytokine tethering. FN fibril assembly is induced by Transforming Growth Factor-β1 (TGF-β1), which was originally identified for its role in malignant transformation. Addition of exogenous TGF-β1 drives FN fibril assembly while also upregulating endogenous TGF-β1 expression and autocrine signaling. In the current study, we sought to determine if autocrine TGF-β1 signaling plays a role in FN fibril formation in either MCF10A mammary epithelial cells, which behave similarly to healthy epithelia, or malignant MDA- MB-231 breast cancer cells. Our results show two interesting findings: first, malignant MDA-MB- 231 cells assemble less FN into fibrils, despite expressing and secreting more soluble FN; second, autocrine TGF-β1 signaling is required for FN fibril formation in MCF10A epithelial cells, even in the presence of exogenous, active TGF-β1. This suggests that autocrine TGF-β1 is signaling through distinct pathways from active exogenous TGF-β1. We hypothesized that this signaling was mediated by interactions between the TGF-β1 latency associated peptide (LAP) and αv integrins; indeed, incubating MCF10As with soluble LAP, even in the absence of the active TGF-β1 ligand, partially recovered FN fibril assembly. Taken together, these data suggests that autocrine TGF-β1 plays a critical role in FN fibril assembly, and this interaction is mediated by LAP-integrin signaling.
Collapse
Affiliation(s)
- Michael M Sofroniou
- Department of Biomedical Engineering, Virginia Commonwealth University, 410 West Main St., Richmond, VA 23284, USA
| | - Christopher A Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, 410 West Main St., Richmond, VA 23284, USA.
| |
Collapse
|
5
|
Rao C, Lin W, Song Z. Analytical refractory period distribution for a class of time-variant biochemical systems with second-order reactions. J Chem Phys 2023; 159:124105. [PMID: 38127379 DOI: 10.1063/5.0156276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/05/2023] [Indexed: 12/23/2023] Open
Abstract
Refractory period (RP), the waiting time between signals, can induce complex signaling dynamics, such as acceleration, adaptation, and oscillation, within many cellular biochemical networks. However, its underlying molecular mechanisms are still unclear. Rigorously estimating the RP distribution may be essential to identify its causal regulatory mechanisms. Traditional methods of estimating the RP distribution depend on solving the underlying Chemical Master Equations (CMEs), the dominant modeling formalism of biochemical systems. However, exact solutions of the CME are only known for simple reaction systems with zero- and first-order reactions or specific systems with second-order reactions. General solutions still need to be derived for systems with bimolecular reactions. It is even more challenging if large state-space and nonconstant reaction rates are involved. Here, we developed a direct method to gain the analytical RP distribution for a class of second-order reaction systems with nonconstant reaction rates and large state space. Instead of using the CME, we used an equivalent path-wise representation, which is the solution to a transformed martingale problem of the CME. This allowed us to bypass solving a CME. We then applied the method to derive the analytical RP distribution of a real complex biochemical network with second-order reactions, the Drosophila phototransduction cascade. Our approach provides an alternative to the CMEs in deriving the analytical RP distributions of a class of second-order reaction systems. Since the bimolecular reactions are common in biological systems, our approach could enhance understanding real-world biochemical processes.
Collapse
Affiliation(s)
- Changqian Rao
- School of Mathematical Sciences and Shanghai Center for Mathematical Sciences, Fudan University, 200433 Shanghai, China
- Research Institute of Intelligent Complex Systems, Fudan University, 200433 Shanghai, China
| | - Wei Lin
- School of Mathematical Sciences and Shanghai Center for Mathematical Sciences, Fudan University, 200433 Shanghai, China
- Research Institute of Intelligent Complex Systems, Fudan University, 200433 Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433 Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, 200032 Shanghai, China
| | - Zhuoyi Song
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 200433 Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, 200032 Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| |
Collapse
|
6
|
Soomro A, Khajehei M, Li R, O’Neil K, Zhang D, Gao B, MacDonald M, Kakoki M, Krepinsky JC. A therapeutic target for CKD: activin A facilitates TGFβ1 profibrotic signaling. Cell Mol Biol Lett 2023; 28:10. [PMID: 36717814 PMCID: PMC9885651 DOI: 10.1186/s11658-023-00424-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND TGFβ1 is a major profibrotic mediator in chronic kidney disease (CKD). Its direct inhibition, however, is limited by adverse effects. Inhibition of activins, also members of the TGFβ superfamily, blocks TGFβ1 profibrotic effects, but the mechanism underlying this and the specific activin(s) involved are unknown. METHODS Cells were treated with TGFβ1 or activins A/B. Activins were inhibited generally with follistatin, or specifically with neutralizing antibodies or type I receptor downregulation. Cytokine levels, signaling and profibrotic responses were assessed with ELISA, immunofluorescence, immunoblotting and promoter luciferase reporters. Wild-type or TGFβ1-overexpressing mice with unilateral ureteral obstruction (UUO) were treated with an activin A neutralizing antibody. RESULTS In primary mesangial cells, TGFβ1 induces secretion primarily of activin A, which enables longer-term profibrotic effects by enhancing Smad3 phosphorylation and transcriptional activity. This results from lack of cell refractoriness to activin A, unlike that for TGFβ1, and promotion of TGFβ type II receptor expression. Activin A also supports transcription through regulating non-canonical MRTF-A activation. TGFβ1 additionally induces secretion of activin A, but not B, from tubular cells, and activin A neutralization prevents the TGFβ1 profibrotic response in renal fibroblasts. Fibrosis induced by UUO is inhibited by activin A neutralization in wild-type mice. Worsened fibrosis in TGFβ1-overexpressing mice is associated with increased renal activin A expression and is inhibited to wild-type levels with activin A neutralization. CONCLUSIONS Activin A facilitates TGFβ1 profibrotic effects through regulation of both canonical (Smad3) and non-canonical (MRTF-A) signaling, suggesting it may be a novel therapeutic target for preventing fibrosis in CKD.
Collapse
Affiliation(s)
- Asfia Soomro
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Mohammad Khajehei
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Renzhong Li
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Kian O’Neil
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Dan Zhang
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Bo Gao
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Melissa MacDonald
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Masao Kakoki
- grid.410711.20000 0001 1034 1720Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Joan C. Krepinsky
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada ,grid.416721.70000 0001 0742 7355St. Joseph’s Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON L8N 4A6 Canada
| |
Collapse
|
7
|
Sarma U, Ripka L, Anyaegbunam UA, Legewie S. Modeling Cellular Signaling Variability Based on Single-Cell Data: The TGFβ-SMAD Signaling Pathway. Methods Mol Biol 2023; 2634:215-251. [PMID: 37074581 DOI: 10.1007/978-1-0716-3008-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Nongenetic heterogeneity is key to cellular decisions, as even genetically identical cells respond in very different ways to the same external stimulus, e.g., during cell differentiation or therapeutic treatment of disease. Strong heterogeneity is typically already observed at the level of signaling pathways that are the first sensors of external inputs and transmit information to the nucleus where decisions are made. Since heterogeneity arises from random fluctuations of cellular components, mathematical models are required to fully describe the phenomenon and to understand the dynamics of heterogeneous cell populations. Here, we review the experimental and theoretical literature on cellular signaling heterogeneity, with special focus on the TGFβ/SMAD signaling pathway.
Collapse
Affiliation(s)
- Uddipan Sarma
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Lorenz Ripka
- Institute of Molecular Biology (IMB), Mainz, Germany
- Department of Systems Biology, Institute for Biomedical Genetics, University of Stuttgart, Stuttgart, Germany
| | - Uchenna Alex Anyaegbunam
- Institute of Molecular Biology (IMB), Mainz, Germany
- Department of Systems Biology, Institute for Biomedical Genetics, University of Stuttgart, Stuttgart, Germany
| | - Stefan Legewie
- Institute of Molecular Biology (IMB), Mainz, Germany.
- Department of Systems Biology, Institute for Biomedical Genetics, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center for Systems Biology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
8
|
Meecham A, Cutmore LC, Protopapa P, Rigby LG, Marshall JF. Ligand-bound integrin αvβ6 internalisation and trafficking. Front Cell Dev Biol 2022; 10:920303. [PMID: 36092709 PMCID: PMC9448872 DOI: 10.3389/fcell.2022.920303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
The integrin αvβ6 is expressed at low levels in most normal healthy tissue but is very often upregulated in a disease context including cancer and fibrosis. Integrins use endocytosis and trafficking as a means of regulating their surface expression and thus their functions, however little is known of how this process is regulated in the context of αvβ6. As αvβ6 is a major target for the development of therapeutics in cancer and fibrosis, understanding these dynamics is critical in the development of αvβ6-targeted therapies. Following development of a flow cytometry-based assay to measure ligand (A20FMDV2 or LAP)-bound αvβ6 endocytosis, an siRNA screen was performed to identify which genes were responsible for internalising αvβ6. These data identified 15 genes (DNM2, CBLB, DNM3, CBL, EEA1, CLTC, ARFGAP3, CAV1, CYTH2, CAV3, CAV2, IQSEC1, AP2M1, TSG101) which significantly decreased endocytosis, predominantly within dynamin-dependent pathways. Inhibition of these dynamin-dependent pathways significantly reduced αvβ6-dependent migration (αvβ6-specific migration was 547 ± 128 under control conditions, reduced to 225 ± 73 with clathrin inhibition, and 280 ± 51 with caveolin inhibition). Colocalization studies of αvβ6 with endosome markers revealed that up to 6 h post-internalisation of ligand, αvβ6 remains in Rab11-positive endosomes in a perinuclear location, with no evidence of αvβ6 degradation up to 48 h post exposure to A20FMDV2. Additionally, 60% of ligand-bound αvβ6 was recycled back to the surface by 6 h. With studies ongoing using conjugated A20FMDV2 to therapeutically target αvβ6 in cancer and fibrosis, these data have important implications. Binding of A20FMDV2 seemingly removes much of the αvβ6 from the cell membrane, and upon its recycling, a large fraction appears to still be in the ligand-bound state. While these results are observed with A20FMDV2, these data will be of value in the design of αvβ6-specific therapeutics and potentially the types of therapeutic load.
Collapse
Affiliation(s)
- Amelia Meecham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- University of California, San Diego, San Diego, CA, United States
| | - Lauren C. Cutmore
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Pantelitsa Protopapa
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lauren G. Rigby
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
9
|
Kolbe N, Hexemer L, Bammert LM, Loewer A, Lukáčová-Medvid’ová M, Legewie S. Data-based stochastic modeling reveals sources of activity bursts in single-cell TGF-β signaling. PLoS Comput Biol 2022; 18:e1010266. [PMID: 35759468 PMCID: PMC9269928 DOI: 10.1371/journal.pcbi.1010266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 07/08/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cells sense their surrounding by employing intracellular signaling pathways that transmit hormonal signals from the cell membrane to the nucleus. TGF-β/SMAD signaling encodes various cell fates, controls tissue homeostasis and is deregulated in diseases such as cancer. The pathway shows strong heterogeneity at the single-cell level, but quantitative insights into mechanisms underlying fluctuations at various time scales are still missing, partly due to inefficiency in the calibration of stochastic models that mechanistically describe signaling processes. In this work we analyze single-cell TGF-β/SMAD signaling and show that it exhibits temporal stochastic bursts which are dose-dependent and whose number and magnitude correlate with cell migration. We propose a stochastic modeling approach to mechanistically describe these pathway fluctuations with high computational efficiency. Employing high-order numerical integration and fitting to burst statistics we enable efficient quantitative parameter estimation and discriminate models that assume noise in different reactions at the receptor level. This modeling approach suggests that stochasticity in the internalization of TGF-β receptors into endosomes plays a key role in the observed temporal bursting. Further, the model predicts the single-cell dynamics of TGF-β/SMAD signaling in untested conditions, e.g., successfully reflects memory effects of signaling noise and cellular sensitivity towards repeated stimulation. Taken together, our computational framework based on burst analysis, noise modeling and path computation scheme is a suitable tool for the data-based modeling of complex signaling pathways, capable of identifying the source of temporal noise.
Collapse
Affiliation(s)
- Niklas Kolbe
- Institute of Geometry and Practical Mathematics, RWTH Aachen University, Aachen, Germany
| | - Lorenz Hexemer
- Institute of Molecular Biology (IMB), Mainz, Germany
- Department of Systems Biology, Institute for Biomedical Genetics (IBMG), University of Stuttgart, Stuttgart, Germany
| | | | - Alexander Loewer
- Systems Biology of the Stress Response, Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | | | - Stefan Legewie
- Department of Systems Biology, Institute for Biomedical Genetics (IBMG), University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center for Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
10
|
Dynamic Visualization of TGF-β/SMAD3 Transcriptional Responses in Single Living Cells. Cancers (Basel) 2022; 14:cancers14102508. [PMID: 35626109 PMCID: PMC9139966 DOI: 10.3390/cancers14102508] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary How a single cytokine can induce a variety of cellular responses in the same cell or in different cells is a longstanding question. Transforming growth factor β (TGF-β) is a prototypical multifunctional cytokine of which biological responses are highly dependent on in a cellular context. TGF-β signals via intracellular SMAD transcription factors, and the duration and intensity of SMAD activation are key determinants for the responses that are elicited by TGF-β. To visualize the TGF-β signaling kinetics, we developed a dynamic TGF-β/SMAD3 transcriptional reporter using a quickly folded and highly unstable green florescent protein. We demonstrate the specificity and sensitivity of this reporter and its wide application to monitor dynamic TGF-β-induced responses in cells cultured on plastic dishes, and in living animals. This tool allows for the analysis of TGF-β signaling at a single living cell level, and allows for the discovery of dynamic TGF-β SMAD- induced transcriptional responses in multi-step biological processes. Abstract Transforming growth factor-β (TGF-β) signaling is tightly controlled in duration and intensity during embryonic development and in the adult to maintain tissue homeostasis. To visualize the TGF-β/SMAD3 signaling kinetics, we developed a dynamic TGF-β/SMAD3 transcriptional fluorescent reporter using multimerized SMAD3/4 binding elements driving the expression of a quickly folded and highly unstable GFP protein. We demonstrate the specificity and sensitivity of this reporter and its wide application to monitor dynamic TGF-β/SMAD3 transcriptional responses in both 2D and 3D systems in vitro, as well as in vivo, using live-cell and intravital imaging. Using this reporter in B16F10 cells, we observed single cell heterogeneity in response to TGF-β challenge, which can be categorized into early, late, and non-responders. Because of its broad application potential, this reporter allows for new discoveries into how TGF-β/SMAD3-dependent transcriptional dynamics are affected during multistep and reversible biological processes.
Collapse
|
11
|
Wu G, Li Y. TGF-β induced reprogramming and drug resistance in triple-negative breast cells. BMC Pharmacol Toxicol 2022; 23:23. [PMID: 35395809 PMCID: PMC8994282 DOI: 10.1186/s40360-022-00561-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/21/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The development of drug resistance remains to be a major cause of therapeutic failure in breast cancer patients. How drug-sensitive cells first evade drug inhibition to proliferate remains to be fully investigated. METHODS Here we characterized the early transcriptional evolution in response to TGF-β in the human triple-negative breast cells through bioinformatical analysis using a published RNA-seq dataset, for which MCF10A cells were treated with 5 ng/ml TGF-β1 for 0 h, 24 h, 48 h and 72 h, and the RNA-seq were performed in biological duplicates. The protein-protein interaction networks of the differentially expressed genes were constructed. KEGG enrichment analysis, cis-regulatory sequence analysis and Kaplan-Meier analysis were also performed to analyze the cellular reprograming induced by TGF-β and its contribution to the survival probability decline of breast cancer patients. RESULT Transcriptomic analysis revealed that cell growth was severely suppressed by TGF-β in the first 24 h but this anti-proliferate impact attenuated between 48 h and 72 h. The oncogenic actions of TGF-β happened within the same time frame with its anti-proliferative effects. In addition, sustained high expression of several drug resistance markers was observed after TGF-β treatment. We also identified 17 TGF-β induced genes that were highly correlated with the survival probability decline of breast cancer patients. CONCLUSION Together, TGF-β plays an important role in tumorigenesis and the development of drug resistance, which implies potential therapeutic strategies targeting the early-stage TGF-β signaling activities.
Collapse
Affiliation(s)
- Guoyu Wu
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China.
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yuchao Li
- MegaLab, MegaRobo Technologies Co., Ltd, Beijing, China
| |
Collapse
|
12
|
Wu G, Xiu H, Luo H, Ding Y, Li Y. A mathematical model for cell cycle control: graded response or quantized response. Cell Cycle 2022; 21:820-834. [PMID: 35107036 PMCID: PMC8973363 DOI: 10.1080/15384101.2022.2031770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/03/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Cell cycle is an important and complex biological system. A lot of efforts have been put in understanding cell cycle arrest for its vital role in clinical therapies. The cell-cycle-arrest outcomes upon stimulation are complicated. The response could be stringent or relaxed, and graded or quantized. A model fully addressing various cell-cycle-arrest outcomes is to be developed. Here, we developed a mathematical model of cell cycle control incorporating distinct characteristics of various cell-cycle-arrest outcomes. The model can simulate two typical properties of cell cycle arrest, quantized and graded. We also characterized the inheritable quiescence and refractory state, which were crucial in long-term response of the population. Then, we monitored cells respond to multiple stimulations, and the results indicated that cells responded to stimulations with small interval did not induce significantly sustained cell cycle arrest as the existence of refractory state. Our work will benefit fundamental research and make efforts to predicting outcomes of clinical therapeutics.
Collapse
Affiliation(s)
- Guoyu Wu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangdong, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- CONTACT Guoyu Wu
| | - Huiyu Xiu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangdong, China
| | - Haiying Luo
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangdong, China
| | - Yu Ding
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangdong, China
| | - Yuchao Li
- MegaLab, MegaRobo Technologies Co., Ltd, Beijing, China
- Yuchao Li
| |
Collapse
|
13
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
14
|
Ji YR, Cheng CC, Lee AL, Shieh JCC, Wu HJ, Huang APH, Hsu YH, Young TH. Poly(allylguanidine)-Coated Surfaces Regulate TGF-β in Glioblastoma Cells to Induce Apoptosis via NF-κB Pathway Activation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:59400-59410. [PMID: 34846137 DOI: 10.1021/acsami.1c21027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Polycationic biomaterials are currently widely applied in neuronal cell cultures to promote cell adhesion and viability. However, polycations generally have cytotoxic properties that limit their application in the field of biomaterials. In this study, we examined the use of a novel polycation poly(allylguanidine) (PAG), which contains a guanidine group in the side chain and a structure similar to poly(allylamine hydrochloride) (PAH), an example of another commonly used polycation. Our findings showed that exposure to PAG induced apoptosis in glioblastoma (GBM) cells, while exposure to PAH induced necrosis. Compared to control groups, the PAG coating significantly limited the proliferation of GBM8901 in vitro and in vivo. Furthermore, GBM8901 cells exposed to the PAG coating exhibited increased levels of phospho-p65 and phosphor-IκB, implying that GBM8901 cells underwent apoptotic cell death via the NF-κB pathway by the regulation of TGF-β. This result was further confirmed to be consistent with the experimental results from western blot protein analysis and apoptosis/necrosis assays. These findings indicate that the polycation PAG has the potential to not only suppress the proliferation of GBM8901 cancer cells but also improve the neural viability and promote the differentiation of neural stem/precursor cells into mature neurons. In conclusion, biomaterials such as PAG act as extremely potent options for applications in the treatment of pathological conditions such as brain cancer.
Collapse
Affiliation(s)
- You-Ren Ji
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Ching-Chia Cheng
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
| | - An-Li Lee
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
- Division of Plastic Surgery, Department of Surgery, MacKay Memorial Hospital, Taipei 104, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | | | - Hsin-Ju Wu
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Abel Po-Hao Huang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei 100, Taiwan
| | - Yi-Hua Hsu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei 100, Taiwan
| | - Tai-Horng Young
- Department of Biomedical Engineering, National Taiwan University, Taipei 100, Taiwan
- Department of Biomedical Engineering, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
15
|
Li Y, Lee H, Heo WD, Zi Z. Revisiting the Role of TGFβ Receptor Internalization for Smad Signaling: It is Not Required in Optogenetic TGFβ Signaling Systems. Adv Biol (Weinh) 2021; 5:e2101008. [PMID: 34463435 DOI: 10.1002/adbi.202101008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/21/2021] [Indexed: 11/11/2022]
Abstract
Endocytosis is an important process by which many signaling receptors reach their intracellular effectors. Accumulating evidence suggests that internalized receptors play critical roles in triggering cellular signaling, including transforming growth factor β (TGFβ) signaling. Despite intensive studies on the TGFβ pathway over the last decades, the necessity of TGFβ receptor endocytosis for downstream TGFβ signaling responses is a subject of debate. In this study, mathematical modeling and synthetic biology approaches are combined to re-evaluate whether TGFβ receptor internalization is indispensable for inducing Smad signaling. It is found that optogenetic systems with plasma membrane-tethered TGFβ receptors can induce fast and sustained Smad2 activation upon light stimulations. Modeling analysis suggests that endocytosis is precluded for the membrane-anchored optogenetic TGFβ receptors. Therefore, this study provides new evidence to support that TGFβ receptor internalization is not required for Smad2 activation.
Collapse
Affiliation(s)
- Yuchao Li
- Max Planck Institute for Molecular Genetics, Otto Warburg Laboratory, 14195, Berlin, Germany
| | - Heeyoung Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305701, Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305701, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305701, Republic of Korea
| | - Zhike Zi
- Max Planck Institute for Molecular Genetics, Otto Warburg Laboratory, 14195, Berlin, Germany.,German Federal Institute for Risk Assessment, Department of Experimental Toxicology and ZEBET, 10589, Berlin, Germany
| |
Collapse
|
16
|
Ramachandran A, Mehić M, Wasim L, Malinova D, Gori I, Blaszczyk BK, Carvalho DM, Shore EM, Jones C, Hyvönen M, Tolar P, Hill CS. Pathogenic ACVR1 R206H activation by Activin A-induced receptor clustering and autophosphorylation. EMBO J 2021; 40:e106317. [PMID: 34003511 PMCID: PMC8280795 DOI: 10.15252/embj.2020106317] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 11/23/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) and diffuse intrinsic pontine glioma (DIPG) are debilitating diseases that share causal mutations in ACVR1, a TGF-β family type I receptor. ACVR1R206H is a frequent mutation in both diseases. Pathogenic signaling via the SMAD1/5 pathway is mediated by Activin A, but how the mutation triggers aberrant signaling is not known. We show that ACVR1 is essential for Activin A-mediated SMAD1/5 phosphorylation and is activated by two distinct mechanisms. Wild-type ACVR1 is activated by the Activin type I receptors, ACVR1B/C. In contrast, ACVR1R206H activation does not require upstream kinases, but is predominantly activated via Activin A-dependent receptor clustering, which induces its auto-activation. We use optogenetics and live-imaging approaches to demonstrate Activin A-induced receptor clustering and show it requires the type II receptors ACVR2A/B. Our data provide molecular mechanistic insight into the pathogenesis of FOP and DIPG by linking the causal activating genetic mutation to disrupted signaling.
Collapse
Affiliation(s)
- Anassuya Ramachandran
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Merima Mehić
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| | - Laabiah Wasim
- Immune Receptor Activation LaboratoryThe Francis Crick InstituteLondonUK
| | | | - Ilaria Gori
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| | | | - Diana M Carvalho
- Division of Molecular PathologyThe Institute of Cancer ResearchSuttonUK
| | - Eileen M Shore
- Departments of Orthopaedic Surgery and GeneticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Chris Jones
- Division of Molecular PathologyThe Institute of Cancer ResearchSuttonUK
| | - Marko Hyvönen
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Pavel Tolar
- Immune Receptor Activation LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
Division of Infection and ImmunityInstitute of Immunity and TransplantationUniversity CollegeLondonUK
| | - Caroline S Hill
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
17
|
Kostopoulou N, Bellou S, Bagli E, Markou M, Kostaras E, Hyvönen M, Kalaidzidis Y, Papadopoulos A, Chalmantzi V, Kyrkou A, Panopoulou E, Fotsis T, Murphy C. Embryonic stem cells are devoid of macropinocytosis, a trafficking pathway for activin A in differentiated cells. J Cell Sci 2021; 134:jcs246892. [PMID: 34313314 DOI: 10.1242/jcs.246892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Ligand-receptor complexes formed at the plasma membrane are internalised via various endocytic pathways that influence the ultimate signalling output by regulating the selection of interaction partners by the complex along the trafficking route. We report that, in differentiated cells, activin A-receptor complexes are internalised via clathrin-mediated endocytosis (CME) and macropinocytosis (MP), whereas in human embryonic stem cells (hESCs) internalisation occurs via CME. We further show that hESCs are devoid of MP, which becomes functional upon differentiation towards endothelial cells through mesoderm mediators. Our results reveal, for the first time, that MP is an internalisation route for activin A in differentiated cells, and that MP is not active in hESCs and is induced as cells differentiate.
Collapse
Affiliation(s)
- Nikoleta Kostopoulou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Sofia Bellou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Confocal Laser Scanning Microscopy Unit, Network of Research Supporting Laboratories, University of Ioannina, Ioannina, 45110, Greece
| | - Eleni Bagli
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Maria Markou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Eleftherios Kostaras
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Yiannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Angelos Papadopoulos
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Varvara Chalmantzi
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Athena Kyrkou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Ekaterini Panopoulou
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Theodore Fotsis
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Carol Murphy
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham, A118 Aston Webb, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
18
|
Ungefroren H. Autocrine TGF-β in Cancer: Review of the Literature and Caveats in Experimental Analysis. Int J Mol Sci 2021; 22:977. [PMID: 33478130 PMCID: PMC7835898 DOI: 10.3390/ijms22020977] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Autocrine signaling is defined as the production and secretion of an extracellular mediator by a cell followed by the binding of that mediator to receptors on the same cell to initiate signaling. Autocrine stimulation often operates in autocrine loops, a type of interaction, in which a cell produces a mediator, for which it has receptors, that upon activation promotes expression of the same mediator, allowing the cell to repeatedly autostimulate itself (positive feedback) or balance its expression via regulation of a second factor that provides negative feedback. Autocrine signaling loops with positive or negative feedback are an important feature in cancer, where they enable context-dependent cell signaling in the regulation of growth, survival, and cell motility. A growth factor that is intimately involved in tumor development and progression and often produced by the cancer cells in an autocrine manner is transforming growth factor-β (TGF-β). This review surveys the many observations of autocrine TGF-β signaling in tumor biology, including data from cell culture and animal models as well as from patients. We also provide the reader with a critical discussion on the various experimental approaches employed to identify and prove the involvement of autocrine TGF-β in a given cellular response.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany;
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany
| |
Collapse
|
19
|
RAC1B Regulation of TGFB1 Reveals an Unexpected Role of Autocrine TGFβ1 in the Suppression of Cell Motility. Cancers (Basel) 2020; 12:cancers12123570. [PMID: 33260366 PMCID: PMC7760153 DOI: 10.3390/cancers12123570] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022] Open
Abstract
Autocrine transforming growth factor (TGF)β has been implicated in epithelial-mesenchymal transition (EMT) and invasion of several cancers including pancreatic ductal adenocarcinoma (PDAC) as well as triple-negative breast cancer (TNBC). However, the precise mechanism and the upstream inducers or downstream effectors of endogenous TGFB1 remain poorly characterized. In both cancer types, the small GTPase RAC1B inhibits cell motility induced by recombinant human TGFβ1 via downregulation of the TGFβ type I receptor, ALK5, but whether RAC1B also impacts autocrine TGFβ signaling has not yet been studied. Intriguingly, RNA interference-mediated knockdown (RNAi-KD) or CRISPR/Cas-mediated knockout of RAC1B in TGFβ1-secreting PDAC-derived Panc1 cells resulted in a dramatic decrease in secreted bioactive TGFβ1 in the culture supernatants and TGFB1 mRNA expression, while the reverse was true for TNBC-derived MDA-MB-231 cells ectopically expressing RAC1B. Surprisingly, the antibody-mediated neutralization of secreted bioactive TGFβ or RNAi-KD of the endogenous TGFB1 gene, was associated with increased rather than decreased migratory activities of Panc1 and MDA-MB-231 cells, upregulation of the promigratory genes SNAI1, SNAI2 and RAC1, and downregulation of the invasion suppressor genes CDH1 (encoding E-cadherin) and SMAD3. Intriguingly, ectopic re-expression of SMAD3 was able to rescue Panc1 and MDA-MB-231 cells from the TGFB1 KD-induced rise in migratory activity. Together, these data suggest that RAC1B favors synthesis and secretion of autocrine TGFβ1 which in a SMAD3-dependent manner blocks EMT-associated gene expression and cell motility.
Collapse
|
20
|
Ungefroren H, Wellner UF, Keck T, Lehnert H, Marquardt JU. The Small GTPase RAC1B: A Potent Negative Regulator of-and Useful Tool to Study-TGFβ Signaling. Cancers (Basel) 2020; 12:E3475. [PMID: 33266416 PMCID: PMC7700615 DOI: 10.3390/cancers12113475] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
RAC1 and its alternatively spliced isoform, RAC1B, are members of the Rho family of GTPases. Both isoforms are involved in the regulation of actin cytoskeleton remodeling, cell motility, cell proliferation, and epithelial-mesenchymal transition (EMT). Compared to RAC1, RAC1B exhibits a number of distinctive features with respect to tissue distribution, downstream signaling and a role in disease conditions like inflammation and cancer. The subcellular locations and interaction partners of RAC1 and RAC1B vary depending on their activation state, which makes RAC1 and RAC1B ideal candidates to establish cross-talk with cancer-associated signaling pathways-for instance, interactions with signaling by transforming growth factor β (TGFβ), a known tumor promoter. Although RAC1 has been found to promote TGFβ-driven tumor progression, recent observations in pancreatic carcinoma cells surprisingly revealed that RAC1B confers anti-oncogenic properties, i.e., through inhibiting TGFβ-induced EMT. Since then, an unexpected array of mechanisms through which RAC1B cross-talks with TGFβ signaling has been demonstrated. However, rather than being uniformly inhibitory, RAC1B interacts with TGFβ signaling in a way that results in the selective blockade of tumor-promoting pathways, while concomitantly allowing tumor-suppressive pathways to proceed. In this review article, we are going to discuss the specific interactions between RAC1B and TGFβ signaling, which occur at multiple levels and include various components such as ligands, receptors, cytosolic mediators, transcription factors, and extracellular inhibitors of TGFβ ligands.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany;
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, Campus Kiel, University Hospital Schleswig-Holstein, D-24105 Kiel, Germany
| | - Ulrich F. Wellner
- Clinic for Surgery, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany; (U.F.W.); (T.K.)
| | - Tobias Keck
- Clinic for Surgery, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany; (U.F.W.); (T.K.)
| | | | - Jens-Uwe Marquardt
- First Department of Medicine, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany;
| |
Collapse
|
21
|
F M, M V, C B, O G, M M, N E. Development of a microfluidic approach for the real-time analysis of extrinsic TGF-β signalling. Biochem Biophys Res Commun 2020; 532:32-39. [PMID: 32826061 DOI: 10.1016/j.bbrc.2020.07.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022]
Abstract
Autocrine and paracrine signalling are traditionally difficult to study due to the sub-micromolar concentrations involved. This has proven to be especially limiting in the study of embryonic stem cells that rely on such signalling for viability, self-renewal, and proliferation. Microfluidics allows to achieve local concentrations of ligands representative of the in vivo stem cell niche, gaining more precise control over the cell microenvironment, as well as to manipulate ligands availability with high temporal resolution and minimal amount of reagents. Here we developed a microfluidics-based system for monitoring the dynamics of TGF-β pathway activity by means of a SMAD2/3-dependent luciferase reporter. We first validated our system by showing dose-dependent transcriptional activation. We then tested the effects of pulsatile stimulation and delayed inhibition of TGF-β activity on signalling dynamics. Finally, we show that our microfluidic system, unlike conventional culture systems, can detect TGF-β ligands secreted in the conditioned medium from hESCs.
Collapse
Affiliation(s)
- Michielin F
- Department of Industrial Engineering, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine (VIMM), Padova, Italy; Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Vetralla M
- Department of Industrial Engineering, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Bolego C
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Gagliano O
- Department of Industrial Engineering, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Montagner M
- Department of Molecular Medicine, University of Padova, Italy
| | - Elvassore N
- Department of Industrial Engineering, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine (VIMM), Padova, Italy; Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China.
| |
Collapse
|
22
|
Olsen OE, Hella H, Elsaadi S, Jacobi C, Martinez-Hackert E, Holien T. Activins as Dual Specificity TGF-β Family Molecules: SMAD-Activation via Activin- and BMP-Type 1 Receptors. Biomolecules 2020; 10:biom10040519. [PMID: 32235336 PMCID: PMC7225989 DOI: 10.3390/biom10040519] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Activins belong to the transforming growth factor (TGF)-β family of multifunctional cytokines and signal via the activin receptors ALK4 or ALK7 to activate the SMAD2/3 pathway. In some cases, activins also signal via the bone morphogenetic protein (BMP) receptor ALK2, causing activation of the SMAD1/5/8 pathway. In this study, we aimed to dissect how activin A and activin B homodimers, and activin AB and AC heterodimers activate the two main SMAD branches. We compared the activin-induced signaling dynamics of ALK4/7-SMAD2/3 and ALK2-SMAD1/5 in a multiple myeloma cell line. Signaling via the ALK2-SMAD1/5 pathway exhibited greater differences between ligands than signaling via ALK4/ALK7-SMAD2/3. Interestingly, activin B and activin AB very potently activated SMAD1/5, resembling the activation commonly seen with BMPs. As SMAD1/5 was also activated by activins in other cell types, we propose that dual specificity is a general mechanism for activin ligands. In addition, we found that the antagonist follistatin inhibited signaling by all the tested activins, whereas the antagonist cerberus specifically inhibited activin B. Taken together, we propose that activins may be considered dual specificity TGF-β family members, critically affecting how activins may be considered and targeted clinically.
Collapse
Affiliation(s)
- Oddrun Elise Olsen
- Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Hematology, St. Olav’s University Hospital, 7030 Trondheim, Norway
| | - Hanne Hella
- Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Samah Elsaadi
- Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Carsten Jacobi
- Novartis Institutes for BioMedical Research Basel, Musculoskeletal Disease Area, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Erik Martinez-Hackert
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Toril Holien
- Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Hematology, St. Olav’s University Hospital, 7030 Trondheim, Norway
- Correspondence: ; Tel.: +47-924-21-162
| |
Collapse
|
23
|
Miller DSJ, Schmierer B, Hill CS. TGF-β family ligands exhibit distinct signalling dynamics that are driven by receptor localisation. J Cell Sci 2019; 132:jcs234039. [PMID: 31217285 PMCID: PMC6679586 DOI: 10.1242/jcs.234039] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/09/2019] [Indexed: 12/29/2022] Open
Abstract
Growth factor-induced signal transduction pathways are tightly regulated at multiple points intracellularly, but how cells monitor levels of extracellular ligand and translate this information into appropriate downstream responses remains unclear. Understanding signalling dynamics is thus a key challenge in determining how cells respond to external cues. Here, we demonstrate that different TGF-β family ligands, namely activin A and BMP4, signal with distinct dynamics, which differ profoundly from those of TGF-β itself. The signalling dynamics are driven by differences in the localisation and internalisation of receptors for each ligand, which in turn determine the capability of cells to monitor levels of extracellular ligand. By using mathematical modelling, we demonstrate that the distinct receptor behaviours and signalling dynamics observed may be primarily driven by differences in ligand-receptor affinity. Furthermore, our results provide a clear rationale for the different mechanisms of pathway regulation found in vivo for each of these growth factors.
Collapse
Affiliation(s)
- Daniel S J Miller
- Developmental Signalling Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Bernhard Schmierer
- Karolinska Institutet, Department of Medical Biochemistry and Biophysics and SciLifeLab Biomedicum 9B, Solnavägen 9, SE-171 65 Solna, Stockholm, Sweden
| | - Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
24
|
Abstract
A handful of core intercellular signaling pathways play pivotal roles in a broad variety of developmental processes. It has remained puzzling how so few pathways can provide the precision and specificity of cell-cell communication required for multicellular development. Solving this requires us to quantitatively understand how developmentally relevant signaling information is actively sensed, transformed and spatially distributed by signaling pathways. Recently, single cell analysis and cell-based reconstitution, among other approaches, have begun to reveal the 'communication codes' through which information is represented in the identities, concentrations, combinations and dynamics of extracellular ligands. They have also revealed how signaling pathways decipher these features and control the spatial distribution of signaling in multicellular contexts. Here, we review recent work reporting the discovery and analysis of communication codes and discuss their implications for diverse developmental processes.
Collapse
Affiliation(s)
- Pulin Li
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Howard Hughes Medical Institute, Pasadena, CA 91125, USA
| |
Collapse
|
25
|
Quantitative relationships between SMAD dynamics and target gene activation kinetics in single live cells. Sci Rep 2019; 9:5372. [PMID: 30926874 PMCID: PMC6440972 DOI: 10.1038/s41598-019-41870-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/20/2019] [Indexed: 12/22/2022] Open
Abstract
The transduction of extracellular signals through signaling pathways that culminate in a transcriptional response is central to many biological processes. However, quantitative relationships between activities of signaling pathway components and transcriptional output of target genes remain poorly explored. Here we developed a dual bioluminescence imaging strategy allowing simultaneous monitoring of nuclear translocation of the SMAD4 and SMAD2 transcriptional activators upon TGF-β stimulation, and the transcriptional response of the endogenous connective tissue growth factor (ctgf) gene. Using cell lines allowing to vary exogenous SMAD4/2 expression levels, we performed quantitative measurements of the temporal profiles of SMAD4/2 translocation and ctgf transcription kinetics in hundreds of individual cells at high temporal resolution. We found that while nuclear translocation efficiency had little impact on initial ctgf transcriptional activation, high total cellular SMAD4 but not SMAD2 levels increased the probability of cells to exhibit a sustained ctgf transcriptional response. The approach we present here allows time-resolved single cell quantification of transcription factor dynamics and transcriptional responses and thereby sheds light on the quantitative relationship between SMADs and target gene responses.
Collapse
|
26
|
Heemskerk I, Burt K, Miller M, Chhabra S, Guerra MC, Liu L, Warmflash A. Rapid changes in morphogen concentration control self-organized patterning in human embryonic stem cells. eLife 2019; 8:e40526. [PMID: 30829572 PMCID: PMC6398983 DOI: 10.7554/elife.40526] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/14/2019] [Indexed: 12/31/2022] Open
Abstract
During embryonic development, diffusible signaling molecules called morphogens are thought to determine cell fates in a concentration-dependent way. Yet, in mammalian embryos, concentrations change rapidly compared to the time for making cell fate decisions. Here, we use human embryonic stem cells (hESCs) to address how changing morphogen levels influence differentiation, focusing on how BMP4 and Nodal signaling govern the cell-fate decisions associated with gastrulation. We show that BMP4 response is concentration dependent, but that expression of many Nodal targets depends on rate of concentration change. Moreover, in a self-organized stem cell model for human gastrulation, expression of these genes follows rapid changes in endogenous Nodal signaling. Our study shows a striking contrast between the specific ways ligand dynamics are interpreted by two closely related signaling pathways, highlighting both the subtlety and importance of morphogen dynamics for understanding mammalian embryogenesis and designing optimized protocols for directed stem cell differentiation. Editorial note This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Idse Heemskerk
- Department of BiosciencesRice UniversityHoustonUnited States
| | - Kari Burt
- Department of BiosciencesRice UniversityHoustonUnited States
| | - Matthew Miller
- Department of BiosciencesRice UniversityHoustonUnited States
| | - Sapna Chhabra
- Systems, Synthetic and Physical Biology ProgramRice UniversityHoustonUnited States
| | | | - Lizhong Liu
- Department of BiosciencesRice UniversityHoustonUnited States
| | - Aryeh Warmflash
- Department of BiosciencesRice UniversityHoustonUnited States
- Department of BioengineeringRice UniversityHoustonUnited States
| |
Collapse
|
27
|
Miller DSJ, Bloxham RD, Jiang M, Gori I, Saunders RE, Das D, Chakravarty P, Howell M, Hill CS. The Dynamics of TGF-β Signaling Are Dictated by Receptor Trafficking via the ESCRT Machinery. Cell Rep 2018; 25:1841-1855.e5. [PMID: 30428352 PMCID: PMC7615189 DOI: 10.1016/j.celrep.2018.10.056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 08/03/2018] [Accepted: 10/15/2018] [Indexed: 01/17/2023] Open
Abstract
Signal transduction pathways stimulated by secreted growth factors are tightly regulated at multiple levels between the cell surface and the nucleus. The trafficking of cell surface receptors is emerging as a key step for regulating appropriate cellular responses, with perturbations in this process contributing to human diseases, including cancer. For receptors recognizing ligands of the transforming growth factor β (TGF-β) family, little is known about how trafficking is regulated or how this shapes signaling dynamics. Here, using whole genome small interfering RNA (siRNA) screens, we have identified the ESCRT (endosomal sorting complex required for transport) machinery as a crucial determinant of signal duration. Downregulation of ESCRT components increases the outputs of TGF-β signaling and sensitizes cells to low doses of ligand in their microenvironment. This sensitization drives an epithelial-to-mesenchymal transition (EMT) in response to low doses of ligand, and we demonstrate a link between downregulation of the ESCRT machinery and cancer survival.
Collapse
Affiliation(s)
- Daniel S J Miller
- Developmental Signalling Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Robert D Bloxham
- Developmental Signalling Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ming Jiang
- High Throughput Screening Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ilaria Gori
- Developmental Signalling Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rebecca E Saunders
- High Throughput Screening Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Debipriya Das
- Developmental Signalling Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Probir Chakravarty
- Bioinformatics and Biostatistics Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Howell
- High Throughput Screening Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
28
|
Abstract
One challenge in biology is to make sense of the complexity of biological networks. A good system to approach this is signaling pathways, whose well-characterized molecular details allow us to relate the internal processes of each pathway to their input-output behavior. In this study, we analyzed mathematical models of three metazoan signaling pathways: the canonical Wnt, MAPK/ERK, and Tgfβ pathways. We find an unexpected convergence: the three pathways behave in some physiological contexts as linear signal transmitters. Testing the results experimentally, we present direct measurements of linear input-output behavior in the Wnt and ERK pathways. Analytics from each model further reveal that linearity arises through different means in each pathway, which we tested experimentally in the Wnt and ERK pathways. Linearity is a desired property in engineering where it facilitates fidelity and superposition in signal transmission. Our findings illustrate how cells tune different complex networks to converge on the same behavior.
Collapse
Affiliation(s)
- Harry Nunns
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Lea Goentoro
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| |
Collapse
|
29
|
Rosengren T, Larsen LJ, Pedersen LB, Christensen ST, Møller LB. TSC1 and TSC2 regulate cilia length and canonical Hedgehog signaling via different mechanisms. Cell Mol Life Sci 2018; 75:2663-2680. [PMID: 29396625 PMCID: PMC6003990 DOI: 10.1007/s00018-018-2761-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 01/03/2018] [Accepted: 01/24/2018] [Indexed: 01/22/2023]
Abstract
Primary cilia are sensory organelles that coordinate multiple cellular signaling pathways, including Hedgehog (HH), Wingless/Int (WNT) and Transforming Growth Factor-β (TGF-β) signaling. Similarly, primary cilia have been implicated in regulation of mTOR signaling, in which Tuberous Sclerosis Complex proteins 1 and 2 (TSC1/2) negatively regulate protein synthesis by inactivating the mTOR complex 1 (mTORC1) at energy limiting states. Here we report that TSC1 and TSC2 regulate Smoothened (SMO)-dependent HH signaling in mouse embryonic fibroblasts (MEFs). Reduced SMO-dependent expression of Gli1 was demonstrated in both Tsc1-/- and Tsc2-/- cells, and we found that Tsc1 is required for TGF-β induced phosphorylation of SMAD2/3 and subsequent expression of the HH signaling effector and transcription factor GLI2. Hedgehog signaling was restored in Tsc1-/- cells after exogenous expression of Gli2, whereas rapamycin restored HH signaling in Tsc2-/- cells. Furthermore, we observed that Tsc1-/- MEFs display significantly elongated cilia, whereas cilia in Tsc2-/- MEFs were shorter than normal. The elongated cilium phenotype of Tsc1-/- MEFs is likely due to increased mTORC1-dependent autophagic flux observed in these cells, as both the autophagic flux and the cilia length phenotype was restored by rapamycin. In addition, ciliary length control in Tsc1-/- MEFs was also influenced by reduced expression of Gli2, which compromised expression of Wnt5a that normally promotes cilia disassembly. In summary, our results support distinct functions of Tsc1 and Tsc2 in cellular signaling as the two genes affect ciliary length control and HH signaling via different mechanisms.
Collapse
Affiliation(s)
- Thomas Rosengren
- Applied Human Molecular Genetics, Clinical Genetic Clinic, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Gl. Landevej 7, 2600, Glostrup, Denmark
| | - Lasse Jonsgaard Larsen
- Applied Human Molecular Genetics, Clinical Genetic Clinic, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Gl. Landevej 7, 2600, Glostrup, Denmark
| | - Lotte Bang Pedersen
- Department of Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - Søren Tvorup Christensen
- Department of Biology, The August Krogh Building, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - Lisbeth Birk Møller
- Applied Human Molecular Genetics, Clinical Genetic Clinic, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Gl. Landevej 7, 2600, Glostrup, Denmark.
| |
Collapse
|
30
|
Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Rosales-Alvarez RE, Macías-Silva M. Transcriptional cofactors Ski and SnoN are major regulators of the TGF-β/Smad signaling pathway in health and disease. Signal Transduct Target Ther 2018; 3:15. [PMID: 29892481 PMCID: PMC5992185 DOI: 10.1038/s41392-018-0015-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/16/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-β signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-β signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-β/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-β signaling outcome as they control the magnitude and duration of the TGF-β signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-β pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer. Proteins that repress molecular signaling through the transforming growth factor-beta (TGF-β) pathway offer promising targets for treating cancer and fibrosis. Marina Macías-Silva and colleagues from the National Autonomous University of Mexico in Mexico City review the ways in which a pair of proteins, called Ski and SnoN, interact with downstream mediators of TGF-β to inhibit the effects of this master growth factor. Aberrant levels of Ski and SnoN have been linked to diverse range of diseases involving cell proliferation run amok, and therapies that regulate the expression of these proteins could help normalize TGF-β signaling to healthier physiological levels. For decades, drug companies have tried to target the TGF-β pathway, with limited success. Altering the activity of these repressors instead could provide a roundabout way of remedying pathogenic TGF-β activity in fibrosis and oncology.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- 1Instituto de Investigaciones Biomédicas at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Diana G Ríos-López
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | | | - Reyna E Rosales-Alvarez
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Marina Macías-Silva
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| |
Collapse
|
31
|
Network and structure based inference of functional single nucleotide polymorphisms associated with the TGFβ1 gene and its role in colorectal cancer (CRC). GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2018.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Zhang J, Tian XJ, Chen YJ, Wang W, Watkins S, Xing J. Pathway crosstalk enables cells to interpret TGF-β duration. NPJ Syst Biol Appl 2018; 4:18. [PMID: 29872541 PMCID: PMC5972147 DOI: 10.1038/s41540-018-0060-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/28/2018] [Accepted: 05/07/2018] [Indexed: 02/07/2023] Open
Abstract
The detection and transmission of the temporal quality of intracellular and extracellular signals is an essential cellular mechanism. It remains largely unexplored how cells interpret the duration information of a stimulus. In this paper, we performed an integrated quantitative and computational analysis on TGF-β induced activation of SNAIL1, a key transcription factor that regulates several subsequent cell fate decisions such as apoptosis and epithelial-to-mesenchymal transition. We demonstrate that crosstalk among multiple TGF-β activated pathways forms a relay from SMAD to GLI1 that initializes and maintains SNAILl expression, respectively. SNAIL1 functions as a key integrator of information from TGF-β signaling distributed through upstream divergent pathways. The intertwined network serves as a temporal checkpoint, so that cells can generate a transient or sustained expression of SNAIL1 depending on TGF-β duration. Furthermore, we observed that TGF-β treatment leads to an unexpected accumulation of GSK3 molecules in an enzymatically active tyrosine phosphorylation form in Golgi apparatus and ER, followed by accumulation of GSK3 molecules in an enzymatically inhibitive serine phosphorylation in the nucleus. Subsequent model analysis and inhibition experiments revealed that the initial localized increase of GSK3 enzymatic activity couples to the positive feedback loop of the substrate Gli1 to form a network motif with multi-objective functions. That is, the motif is robust against stochastic fluctuations, and has a narrow distribution of response time that is insensitive to initial conditions. Specifically for TGF-β signaling, the motif ensures a smooth relay from SMAD to GLI1 on regulating SNAIL1 expression.
Collapse
Affiliation(s)
- Jingyu Zhang
- 1Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Xiao-Jun Tian
- 1Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA.,4Present Address: School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287 USA
| | - Yi-Jiun Chen
- 1Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Weikang Wang
- 1Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Simon Watkins
- 2Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Jianhua Xing
- 1Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA.,3UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232 USA
| |
Collapse
|
33
|
Nodal and BMP dispersal during early zebrafish development. Dev Biol 2018; 447:14-23. [PMID: 29653088 DOI: 10.1016/j.ydbio.2018.04.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/30/2022]
Abstract
The secreted TGF-β superfamily signals Nodal and BMP coordinate the patterning of vertebrate embryos. Nodal specifies endoderm and mesoderm during germ layer formation, and BMP specifies ventral fates and patterns the dorsal/ventral axis. Five major models have been proposed to explain how the correct distributions of Nodal and BMP are achieved within tissues to orchestrate embryogenesis: source/sink, transcriptional determination, relay, self-regulation, and shuttling. Here, we discuss recent experiments probing these signal dispersal models, focusing on early zebrafish development.
Collapse
|
34
|
Rose AM, Spender LC, Stephen C, Mitchell A, Rickaby W, Bray S, Evans AT, Dayal J, Purdie KJ, Harwood CA, Proby CM, Leigh IM, Coates PJ, Inman GJ. Reduced SMAD2/3 activation independently predicts increased depth of human cutaneous squamous cell carcinoma. Oncotarget 2018; 9:14552-14566. [PMID: 29581863 PMCID: PMC5865689 DOI: 10.18632/oncotarget.24545] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 02/10/2018] [Indexed: 11/25/2022] Open
Abstract
The incidence of cutaneous squamous cell carcinoma (cSCC) is rising. Whilst the majority are cured surgically, aggressive metastatic cSCC carry a poor prognosis. Inactivating mutations in transforming growth factor beta (TGF-β) receptors have been identified amongst genetic drivers of sporadic tumours and murine models of cSCC, suggesting a tumour suppressor function for TGF-β in normal skin. However, paradoxically, TGF-β acts as a tumour promoter in some murine model systems. Few studies have analysed the role of TGF-β/activin signalling in human normal skin, hyper-proliferative skin disorders and cSCC. Antibodies recognising phospho-SMAD proteins which are activated during canonical TGF-β/activin signalling were validated for use in immunohistochemistry. A tissue microarray comprising FFPE lesional and perilesional tissue from human primary invasive cSCC (n=238), cSCC in-situ (n=2) and keratocanthoma (n=9) were analysed in comparison with tissues from normal human scalp (n=10). Phosphorylated SMAD2 and SMAD3 were detected in normal interfollicular epidermal keratinocytes and were also highly localised to inner root sheath, matrix cells and Keratin 15 positive cells. Lesional cSCC tissue had significantly reduced activated SMAD2/3 compared to perilesional tissue, consistent with a tumour suppressor role for SMAD2/3 activators in cSCC. Increased cSCC tumour thickness inversely correlated with the presence of phospho-SMADs in tumour tissue suggesting that a reduction in canonical TGF-β/activin signalling may be associated with disease progression.
Collapse
Affiliation(s)
- Aidan M Rose
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, DD1 9SY, UK.,Department of Plastic and Reconstructive Surgery, Ninewells Hospital and Medical School, NHS Tayside, Dundee, Scotland, DD1 9SY, UK
| | - Lindsay C Spender
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, DD1 9SY, UK
| | - Christopher Stephen
- Department of Plastic and Reconstructive Surgery, Ninewells Hospital and Medical School, NHS Tayside, Dundee, Scotland, DD1 9SY, UK
| | - Alastair Mitchell
- Department of Dermatology, Ninewells Hospital and Medical School, NHS Tayside, Dundee, Scotland, DD1 9SY, UK
| | - William Rickaby
- Dermatopathology Laboratory, St. John's Institute of Dermatology, St.Thomas' Hospital, London, SE1 7EH, UK
| | - Susan Bray
- Tayside Tissue Bank, Ninewells Hospital and Medical School, NHS Tayside, Dundee, Scotland, DD1 9SY, UK
| | - Alan T Evans
- Department of Pathology, Ninewells Hospital and Medical School, NHS Tayside, Dundee, Scotland, DD1 9SY, UK
| | - Jasbani Dayal
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, DD1 9SY, UK
| | - Karin J Purdie
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Catherine A Harwood
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Charlotte M Proby
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, DD1 9SY, UK
| | - Irene M Leigh
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, DD1 9SY, UK.,Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Philip J Coates
- Tayside Tissue Bank, Ninewells Hospital and Medical School, NHS Tayside, Dundee, Scotland, DD1 9SY, UK.,Regional Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, 656 53, Czech Republic
| | - Gareth J Inman
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, DD1 9SY, UK
| |
Collapse
|
35
|
Li Y, Lee M, Kim N, Wu G, Deng D, Kim JM, Liu X, Heo WD, Zi Z. Spatiotemporal Control of TGF-β Signaling with Light. ACS Synth Biol 2018; 7:443-451. [PMID: 29241005 DOI: 10.1021/acssynbio.7b00225] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cells employ signaling pathways to make decisions in response to changes in their immediate environment. Transforming growth factor beta (TGF-β) is an important growth factor that regulates many cellular functions in development and disease. Although the molecular mechanisms of TGF-β signaling have been well studied, our understanding of this pathway is limited by the lack of tools that allow the control of TGF-β signaling with high spatiotemporal resolution. Here, we developed an optogenetic system (optoTGFBRs) that enables the precise control of TGF-β signaling in time and space. Using the optoTGFBRs system, we show that TGF-β signaling can be selectively and sequentially activated in single cells through the modulation of the pattern of light stimulations. By simultaneously monitoring the subcellular localization of TGF-β receptor and Smad2 proteins, we characterized the dynamics of TGF-β signaling in response to different patterns of blue light stimulations. The spatial and temporal precision of light control will make the optoTGFBRs system as a powerful tool for quantitative analyses of TGF-β signaling at the single cell level.
Collapse
Affiliation(s)
- Yuchao Li
- Otto-Warburg
Laboratory, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| | - Minji Lee
- Department
of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Nury Kim
- Center
for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Guoyu Wu
- Otto-Warburg
Laboratory, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| | - Difan Deng
- Otto-Warburg
Laboratory, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| | - Jin Man Kim
- Department
of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Xuedong Liu
- Department
of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, Colorado 80309-0596, United States
| | - Won Do Heo
- Department
of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Center
for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
- KAIST
Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Zhike Zi
- Otto-Warburg
Laboratory, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| |
Collapse
|
36
|
Ramachandran A, Vizán P, Das D, Chakravarty P, Vogt J, Rogers KW, Müller P, Hinck AP, Sapkota GP, Hill CS. TGF-β uses a novel mode of receptor activation to phosphorylate SMAD1/5 and induce epithelial-to-mesenchymal transition. eLife 2018; 7:e31756. [PMID: 29376829 PMCID: PMC5832415 DOI: 10.7554/elife.31756] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/26/2018] [Indexed: 12/15/2022] Open
Abstract
The best characterized signaling pathway downstream of transforming growth factor β (TGF-β) is through SMAD2 and SMAD3. However, TGF-β also induces phosphorylation of SMAD1 and SMAD5, but the mechanism of this phosphorylation and its functional relevance is not known. Here, we show that TGF-β-induced SMAD1/5 phosphorylation requires members of two classes of type I receptor, TGFBR1 and ACVR1, and establish a new paradigm for receptor activation where TGFBR1 phosphorylates and activates ACVR1, which phosphorylates SMAD1/5. We demonstrate the biological significance of this pathway by showing that approximately a quarter of the TGF-β-induced transcriptome depends on SMAD1/5 signaling, with major early transcriptional targets being the ID genes. Finally, we show that TGF-β-induced epithelial-to-mesenchymal transition requires signaling via both the SMAD3 and SMAD1/5 pathways, with SMAD1/5 signaling being essential to induce ID1. Therefore, combinatorial signaling via both SMAD pathways is essential for the full TGF-β-induced transcriptional program and physiological responses.
Collapse
Affiliation(s)
| | - Pedro Vizán
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Debipriya Das
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Probir Chakravarty
- Bioinformatics and Biostatistics FacilityThe Francis Crick InstituteLondonUnited Kingdom
| | - Janis Vogt
- Medical Research Council Protein Phosphorylation and Ubiquitylation UnitUniversity of DundeeDundeeUnited Kingdom
| | | | - Patrick Müller
- Friedrich Miescher Laboratory of the Max Planck SocietyTübingenGermany
| | - Andrew P Hinck
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghUnited States
| | - Gopal P Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation UnitUniversity of DundeeDundeeUnited Kingdom
| | - Caroline S Hill
- Developmental Signalling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
37
|
Strasen J, Sarma U, Jentsch M, Bohn S, Sheng C, Horbelt D, Knaus P, Legewie S, Loewer A. Cell-specific responses to the cytokine TGFβ are determined by variability in protein levels. Mol Syst Biol 2018; 14:e7733. [PMID: 29371237 PMCID: PMC5787704 DOI: 10.15252/msb.20177733] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The cytokine TGFβ provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFβ are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFβ is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity.
Collapse
Affiliation(s)
- Jette Strasen
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center in the Helmholtz Association, Berlin, Germany
| | - Uddipan Sarma
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Marcel Jentsch
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center in the Helmholtz Association, Berlin, Germany.,Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Stefan Bohn
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Caibin Sheng
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center in the Helmholtz Association, Berlin, Germany.,Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Daniel Horbelt
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Alexander Loewer
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center in the Helmholtz Association, Berlin, Germany .,Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
38
|
Martinez Guimera A, Welsh CM, Proctor CJ, McArdle A, Shanley DP. 'Molecular habituation' as a potential mechanism of gradual homeostatic loss with age. Mech Ageing Dev 2017; 169:53-62. [PMID: 29146308 PMCID: PMC5846846 DOI: 10.1016/j.mad.2017.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/26/2017] [Accepted: 11/10/2017] [Indexed: 12/17/2022]
Abstract
Constitutive signals indicate homeostatic dysregulation but their effect on signal transduction remains largely unexplored. A theoretical approach is undertaken to examine how oxidative stress may affect redox signal transduction. Constitutive signals can result in a ‘molecular habituation’ effect that interferes with information transmission. The robustness of such a theoretical observation to the underlying methodology hints at the generality of this principle.
The ability of reactive oxygen species (ROS) to cause molecular damage has meant that chronic oxidative stress has been mostly studied from the point of view of being a source of toxicity to the cell. However, the known duality of ROS molecules as both damaging agents and cellular redox signals implies another perspective in the study of sustained oxidative stress. This is a perspective of studying oxidative stress as a constitutive signal within the cell. In this work, we adopt a theoretical perspective as an exploratory and explanatory approach to examine how chronic oxidative stress can interfere with signal processing by redox signalling pathways in the cell. We report that constitutive signals can give rise to a ‘molecular habituation’ effect that can prime for a gradual loss of biological function. This is because a constitutive signal in the environment has the potential to reduce the responsiveness of a signalling pathway through the prolonged activation of negative regulators. Additionally, we demonstrate how this phenomenon is likely to occur in different signalling pathways exposed to persistent signals and furthermore at different levels of biological organisation.
Collapse
Affiliation(s)
- Alvaro Martinez Guimera
- Institute for Cell and Molecular Biosciences (ICaMB), Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL,United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Ciaran M Welsh
- Institute for Cell and Molecular Biosciences (ICaMB), Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL,United Kingdom
| | - Carole J Proctor
- Institute of Cellular Medicine, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL, United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Anne McArdle
- Department of Musculoskeletal Biology, University of Liverpool (University, Not-for-profit), Institute of Ageing and Chronic Disease,William Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Daryl P Shanley
- Institute for Cell and Molecular Biosciences (ICaMB), Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL,United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom.
| |
Collapse
|
39
|
Khatibi S, Babon J, Wagner J, Manton JH, Tan CW, Zhu HJ, Wormald S, Burgess AW. TGF-β and IL-6 family signalling crosstalk: an integrated model. Growth Factors 2017; 35:100-124. [PMID: 28948853 DOI: 10.1080/08977194.2017.1363746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mathematical models for TGF-β and IL-6 signalling have been linked, providing a platform for analyzing the crosstalk between the systems. An integrated IL-6:TGF-β model was developed via a reduced set of reaction equations which incorporate both feedback loops and appropriate time-delays for transcription and translation processes. The model simulates stable, robust and realistic responses to both ligands. Pulsatile (multiple pulses) inputs for both TGF-β and IL-6 have been simulated to investigate the effects of each ligand on the sensitivity, equilibrium and dynamic responses of the integrated signalling system. In our simulations the crosstalk between constant IL-6 and TGF-β signalling via SMAD7 does not appear to be sufficient to render the cells resistant to TGF-β inhibition. However, the simulations predict that pulsatile IL-6 stimulation would increase SMAD7 levels substantially and consequentially, lead to resistance to TGF-β. The model also allows the prediction of the integrated signalling pathway responses to the mutation of key components, e.g. Gp130 F/F.
Collapse
Affiliation(s)
- Shabnam Khatibi
- a Department of Electrical and Electronic Engineering , University of Melbourne , Parkville , VIC , Australia
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
| | - Jeff Babon
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
| | - John Wagner
- a Department of Electrical and Electronic Engineering , University of Melbourne , Parkville , VIC , Australia
- c IBM Researchtreetience , Carlton , Australia
- d Department of Medical Biology , University of Melbourne , Parkville , VIC , Australia
| | - Jonathan H Manton
- a Department of Electrical and Electronic Engineering , University of Melbourne , Parkville , VIC , Australia
| | - Chin Wee Tan
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
- e IBM Research Collaboratory for Life Sciences Research , Victorian Life Sciences Computation Initiative , Carlton , VIC , Australia
| | - Hong-Jian Zhu
- f Department of Surgery (RMH) , University of Melbourne , Parkville , VIC , Australia
| | - Sam Wormald
- g Division of Cancer and Haematology , The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
| | - Antony W Burgess
- b Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research (WEHI) , Parkville , VIC , Australia
- e IBM Research Collaboratory for Life Sciences Research , Victorian Life Sciences Computation Initiative , Carlton , VIC , Australia
| |
Collapse
|
40
|
Kumar R, Gont A, Perkins TJ, Hanson JEL, Lorimer IAJ. Induction of senescence in primary glioblastoma cells by serum and TGFβ. Sci Rep 2017; 7:2156. [PMID: 28526854 PMCID: PMC5438350 DOI: 10.1038/s41598-017-02380-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/18/2017] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma is the most common type of adult brain tumour and has a median survival after diagnosis of a little more than a year. Glioblastomas have a high frequency of mutations in the TERT promoter and CDKN2A locus that are expected to render them resistant to both replicative and oncogene-induced senescence. However, exposure of PriGO8A primary glioblastoma cells to media with 10% serum induced a senescence-like phenotype characterized by increased senescence-associated β galactosidase activity, PML bodies and p21 and morphological changes typical of senescence. Microarray expression analysis showed that 24 h serum exposure increased the expression of genes associated with the TGFβ pathway. Treatment of PriGO8A cells with TGFβ was sufficient to induce senescence in these cells. The response of PriGO8A cells to serum was dependent on basal expression of the TGFβ activator protein thrombospondin. Primary glioblastoma cells from three additional patients showed a variable ability to undergo senescence in response to serum. However all were able to undergo senescence in response to TGFβ, although for cells from one patient this required concomitant inhibition of Ras pathway signalling. Primary glioblastoma cells therefore retain a functional senescence program that is inducible by acute activation of the TGFβ signalling pathway.
Collapse
Affiliation(s)
- Ritesh Kumar
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexander Gont
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Theodore J Perkins
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Jennifer E L Hanson
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | - Ian A J Lorimer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada. .,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada. .,Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
41
|
Fan X, Gaur U, Sun L, Yang D, Yang M. The Growth Differentiation Factor 11 (GDF11) and Myostatin (MSTN) in tissue specific aging. Mech Ageing Dev 2017; 164:108-112. [PMID: 28472635 DOI: 10.1016/j.mad.2017.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 04/18/2017] [Accepted: 04/28/2017] [Indexed: 01/24/2023]
Abstract
Growth differentiation factor 11 (GDF11) and myostatin (MSTN) are evolutionarily conserved homologues proteins which are closely related members of the transforming growth factor β superfamily. They are often perceived to serve similar or overlapping roles. Recently, GDF11 has been identified as playing a role during aging, however there are conflicting reports as to the nature of this role. In this review, we will discuss the literature regarding functions of GDF11 and myostatin in the heart, brain, and skeletal muscle during aging. Consequently we expect to develop a deeper understanding about the function of these two proteins in organismal aging and disease.
Collapse
Affiliation(s)
- Xiaolan Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University Chengdu, 611130, PR China
| | - Uma Gaur
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University Chengdu, 611130, PR China
| | - Lin Sun
- Jiangsu Vocational College of Medicine, Yancheng, 224000, PR China
| | - Deying Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University Chengdu, 611130, PR China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University Chengdu, 611130, PR China.
| |
Collapse
|
42
|
Ansa-Addo EA, Zhang Y, Yang Y, Hussey GS, Howley BV, Salem M, Riesenberg B, Sun S, Rockey DC, Karvar S, Howe PH, Liu B, Li Z. Membrane-organizing protein moesin controls Treg differentiation and antitumor immunity via TGF-β signaling. J Clin Invest 2017; 127:1321-1337. [PMID: 28287407 DOI: 10.1172/jci89281] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 01/17/2017] [Indexed: 12/22/2022] Open
Abstract
Moesin is a member of the ezrin-radixin-moesin (ERM) family of proteins that are important for organizing membrane domains and receptor signaling and regulating the migration of effector T cells. Whether moesin plays any role during the generation of TGF-β-induced Tregs (iTregs) is unknown. Here, we have discovered that moesin is translationally regulated by TGF-β and is also required for optimal TGF-β signaling that promotes efficient development of iTregs. Loss of moesin impaired the development and function of both peripherally derived iTregs and in vitro-induced Tregs. Mechanistically, we identified an interaction between moesin and TGF-β receptor II (TβRII) that allows moesin to control the surface abundance and stability of TβRI and TβRII. We also found that moesin is required for iTreg conversion in the tumor microenvironment, and the deletion of moesin from recipient mice supported the rapid expansion of adoptively transferred CD8+ T cells against melanoma. Our study establishes moesin as an important regulator of the surface abundance and stability of TβRII and identifies moesin's role in facilitating the efficient generation of iTregs. It also provides an advancement to our understanding about the role of the ERM proteins in regulating signal transduction pathways and suggests that modulation of moesin is a potential therapeutic target for Treg-related immune disorders.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Cell Differentiation
- Cell Membrane/metabolism
- Cells, Cultured
- Female
- HEK293 Cells
- Humans
- Male
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Microfilament Proteins/physiology
- Neoplasm Transplantation
- Protein Binding
- Protein Biosynthesis
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Stability
- Protein Transport
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Skin Neoplasms/therapy
- T-Lymphocytes, Regulatory/physiology
- Transcriptional Activation
- Transforming Growth Factor beta/physiology
- Tumor Escape
- Up-Regulation
Collapse
|
43
|
Britton GJ, Ambler R, Clark DJ, Hill EV, Tunbridge HM, McNally KE, Burton BR, Butterweck P, Sabatos-Peyton C, Hampton-O’Neil LA, Verkade P, Wülfing C, Wraith DC. PKCθ links proximal T cell and Notch signaling through localized regulation of the actin cytoskeleton. eLife 2017; 6:e20003. [PMID: 28112644 PMCID: PMC5310840 DOI: 10.7554/elife.20003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 01/22/2017] [Indexed: 11/16/2022] Open
Abstract
Notch is a critical regulator of T cell differentiation and is activated through proteolytic cleavage in response to ligand engagement. Using murine myelin-reactive CD4 T cells, we demonstrate that proximal T cell signaling modulates Notch activation by a spatiotemporally constrained mechanism. The protein kinase PKCθ is a critical mediator of signaling by the T cell antigen receptor and the principal costimulatory receptor CD28. PKCθ selectively inactivates the negative regulator of F-actin generation, Coronin 1A, at the center of the T cell interface with the antigen presenting cell (APC). This allows for effective generation of the large actin-based lamellum required for recruitment of the Notch-processing membrane metalloproteinase ADAM10. Such enhancement of Notch activation is critical for efficient T cell proliferation and Th17 differentiation. We reveal a novel mechanism that, through modulation of the cytoskeleton, controls Notch activation at the T cell:APC interface thereby linking T cell receptor and Notch signaling pathways.
Collapse
Affiliation(s)
- Graham J Britton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Rachel Ambler
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Danielle J Clark
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Elaine V Hill
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Helen M Tunbridge
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Kerrie E McNally
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Bronwen R Burton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Philomena Butterweck
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | | | - Lea A Hampton-O’Neil
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Paul Verkade
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Christoph Wülfing
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - David Cameron Wraith
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
44
|
Morel PA, Lee REC, Faeder JR. Demystifying the cytokine network: Mathematical models point the way. Cytokine 2016; 98:115-123. [PMID: 27919524 DOI: 10.1016/j.cyto.2016.11.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022]
Abstract
Cytokines provide the means by which immune cells communicate with each other and with parenchymal cells. There are over one hundred cytokines and many exist in families that share receptor components and signal transduction pathways, creating complex networks. Reductionist approaches to understanding the role of specific cytokines, through the use of gene-targeted mice, have revealed further complexity in the form of redundancy and pleiotropy in cytokine function. Creating an understanding of the complex interactions between cytokines and their target cells is challenging experimentally. Mathematical and computational modeling provides a robust set of tools by which complex interactions between cytokines can be studied and analyzed, in the process creating novel insights that can be further tested experimentally. This review will discuss and provide examples of the different modeling approaches that have been used to increase our understanding of cytokine networks. This includes discussion of knowledge-based and data-driven modeling approaches and the recent advance in single-cell analysis. The use of modeling to optimize cytokine-based therapies will also be discussed.
Collapse
Affiliation(s)
- Penelope A Morel
- Department of Immunology, University of Pittsburgh, Pittsburgh, USA.
| | - Robin E C Lee
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, USA
| | - James R Faeder
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, USA
| |
Collapse
|
45
|
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: 62] [Impact Index Per Article: 6.9] [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
|
46
|
Rys JP, DuFort CC, Monteiro DA, Baird MA, Oses-Prieto JA, Chand S, Burlingame AL, Davidson MW, Alliston TN. Discrete spatial organization of TGFβ receptors couples receptor multimerization and signaling to cellular tension. eLife 2015; 4:e09300. [PMID: 26652004 PMCID: PMC4728123 DOI: 10.7554/elife.09300] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 11/04/2015] [Indexed: 11/13/2022] Open
Abstract
Cell surface receptors are central to the cell's ability to generate coordinated responses to the multitude of biochemical and physical cues in the microenvironment. However, the mechanisms by which receptors enable this concerted cellular response remain unclear. To investigate the effect of cellular tension on cell surface receptors, we combined novel high-resolution imaging and single particle tracking with established biochemical assays to examine TGFβ signaling. We find that TGFβ receptors are discretely organized to segregated spatial domains at the cell surface. Integrin-rich focal adhesions organize TβRII around TβRI, limiting the integration of TβRII while sequestering TβRI at these sites. Disruption of cellular tension leads to a collapse of this spatial organization and drives formation of heteromeric TβRI/TβRII complexes and Smad activation. This work details a novel mechanism by which cellular tension regulates TGFβ receptor organization, multimerization, and function, providing new insight into the mechanisms that integrate biochemical and physical cues. DOI:http://dx.doi.org/10.7554/eLife.09300.001 Cells constantly encounter diverse physical and biological signals in their surroundings. Information contained in these signals is transmitted from the cell surface to the interior to trigger coordinated changes in the cell’s behavior. Physical signals include the forces generated by cells pulling on one another or on their surroundings. These pulling forces calibrate the cell’s response to biological signals through mechanisms that remain unclear. The cell surface contains many different proteins that are specialized to sense these signals and guide the cell’s response. In animals, these membrane proteins include the receptors that detect a small signaling protein known as TGFβ. TGFβ first binds to one of these receptors (called TβRII). Next another receptor (called TβRI) is recruited to the complex. Once this complex is formed, the TGFβ receptors activate a complicated signaling pathway that controls how cells grow and divide. Previous work has shown that the TGFβ pathway can also sense and respond to mechanical forces. But it remains poorly understood how pulling forces (or tension) impact TGFβ receptors at the cell surface. Rys, DuFort et al. have now used cutting-edge microscopy and biochemical techniques to analyze individual TβRI and TβRII receptors and observe how they respond to mechanical forces in real-time. This revealed that TβRI and TβRII exist in discrete regions on the cell surface. Rys, DuFort et al. observed that TβRI is enriched at assemblies of molecules called focal adhesions. Focal adhesions are the sites on cell surfaces that allow cells to adhere to one another and to the molecular scaffolding in their surroundings. Unlike TβRI, TβRII was often excluded from these sites and more commonly appeared to ‘bounce’ around the edges of individual focal adhesions. Therefore, focal adhesions limit the interactions between TβRI and TβRII, by sequestering one away from the other. Rys, DuFort et al. next treated cells with a chemical that disrupts tension, and saw that the physical separation between TβRI and TβRII collapsed, which permitted these two receptors to interact and form a working signaling complex. Further work is needed to understand how physical control of TGFβ receptor interactions helps cells coordinate their tasks in response to the myriad biological and physical signals in their surroundings. DOI:http://dx.doi.org/10.7554/eLife.09300.002
Collapse
Affiliation(s)
- Joanna P Rys
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, United States.,Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Christopher C DuFort
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - David A Monteiro
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, United States.,Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Michelle A Baird
- National High Magnetic Field Laboratory,Department of Biological Science, Florida State University, Tallahassee, United States
| | - Juan A Oses-Prieto
- Mass Spectrometry Facility, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Shreya Chand
- Mass Spectrometry Facility, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Alma L Burlingame
- Mass Spectrometry Facility, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Michael W Davidson
- National High Magnetic Field Laboratory,Department of Biological Science, Florida State University, Tallahassee, United States
| | - Tamara N Alliston
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, United States.,Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States.,Department of Bioengineering and Therapeutic Sciences, Department of Otolaryngology-Head and Neck Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
47
|
van Boxtel AL, Chesebro JE, Heliot C, Ramel MC, Stone RK, Hill CS. A Temporal Window for Signal Activation Dictates the Dimensions of a Nodal Signaling Domain. Dev Cell 2015; 35:175-85. [PMID: 26506307 PMCID: PMC4640439 DOI: 10.1016/j.devcel.2015.09.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 08/11/2015] [Accepted: 09/23/2015] [Indexed: 11/22/2022]
Abstract
Morphogen signaling is critical for the growth and patterning of tissues in embryos and adults, but how morphogen signaling gradients are generated in tissues remains controversial. The morphogen Nodal was proposed to form a long-range signaling gradient via a reaction-diffusion system, on the basis of differential diffusion rates of Nodal and its antagonist Lefty. Here we use a specific zebrafish Nodal biosensor combined with immunofluorescence for phosphorylated Smad2 to demonstrate that endogenous Nodal is unlikely to diffuse over a long range. Instead, short-range Nodal signaling activation in a temporal window is sufficient to determine the dimensions of the Nodal signaling domain. The size of this temporal window is set by the differentially timed production of Nodal and Lefty, which arises mainly from repression of Lefty translation by the microRNA miR-430. Thus, temporal information is transformed into spatial information to define the dimensions of the Nodal signaling domain and, consequently, to specify mesendoderm.
Collapse
Affiliation(s)
- Antonius L van Boxtel
- Developmental Signalling, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - John E Chesebro
- Developmental Signalling, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Claire Heliot
- Developmental Signalling, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Marie-Christine Ramel
- Developmental Signalling, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Richard K Stone
- Experimental Histopathology, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Caroline S Hill
- Developmental Signalling, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK.
| |
Collapse
|
48
|
Warsinske HC, Ashley SL, Linderman JJ, Moore BB, Kirschner DE. Identifying Mechanisms of Homeostatic Signaling in Fibroblast Differentiation. Bull Math Biol 2015; 77:1556-82. [PMID: 26384829 DOI: 10.1007/s11538-015-0096-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/21/2015] [Indexed: 10/23/2022]
Abstract
Fibroblasts play an important role in the wound-healing process by generating extracellular matrix (ECM) and undergoing differentiation into myofibroblasts, but these cells can also be involved in pathologic remodeling of tissue. Nascent ECM provides a substrate for re-epithelialization to occur, restoring damaged tissue to a functional state. Dysregulation of this process can result in fibrosis--stiffening and scarring of the tissue. Current treatments cannot halt or reverse this process. The molecular mechanisms underlying fibrotic dysregulation are poorly understood, providing an untapped pool of potential therapeutic targets. Transforming growth factor-β (TGF-β) and adhesion signaling are involved in inducing fibroblast differentiation into α-smooth muscle actin (αSMA) expressing myofibroblasts, while prostaglandin E₂ (PGE₂) has been shown to antagonize TGF-β signaling; however, the temporal and mechanistic details of this relationship have not yet been fully characterized. We measured αSMA, a marker of fibroblast to myofibroblast differentiation, as a function of: TGF-β1 receptor-ligand complex internalization, PGE₂ binding, and adhesion signaling and developed a mathematical model capturing the molecular mechanisms of fibroblast differentiation. Using our model, we predict the following: Periodic dosing with PGE₂ temporarily renders fibroblasts incapable of differentiation and refractory to additional TGF-β1 stimulation; conversely, periodic dosing with TGF-β1 in the presence of PGE₂ induces a reduced signal response that can be further inhibited by the addition of more PGE₂. Controlled fibroblast differentiation is necessary for effective wound healing; however, excessive accumulation of αSMA-expressing myofibroblasts can result in fibrosis. Homeostasis of αSMA in our model requires a balance of positive and negative regulatory signals. Sensitivity analysis predicts that PGE₂ availability, TGF-β1 availability, and the rate of TGF-β1 receptor recycling each highly influence the rates of αSMA production. With this model, we are able to demonstrate that regulation of both TGF-β1 and PGE₂ signaling levels is essential for preventing fibroblast dysregulation.
Collapse
Affiliation(s)
- Hayley C Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shanna L Ashley
- Immunology Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | | | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA, Slapak CA, Lahn MM. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015; 9:4479-99. [PMID: 26309397 PMCID: PMC4539082 DOI: 10.2147/dddt.s86621] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) signaling regulates a wide range of biological processes. TGF-β plays an important role in tumorigenesis and contributes to the hallmarks of cancer, including tumor proliferation, invasion and metastasis, inflammation, angiogenesis, and escape of immune surveillance. There are several pharmacological approaches to block TGF-β signaling, such as monoclonal antibodies, vaccines, antisense oligonucleotides, and small molecule inhibitors. Galunisertib (LY2157299 monohydrate) is an oral small molecule inhibitor of the TGF-β receptor I kinase that specifically downregulates the phosphorylation of SMAD2, abrogating activation of the canonical pathway. Furthermore, galunisertib has antitumor activity in tumor-bearing animal models such as breast, colon, lung cancers, and hepatocellular carcinoma. Continuous long-term exposure to galunisertib caused cardiac toxicities in animals requiring adoption of a pharmacokinetic/pharmacodynamic-based dosing strategy to allow further development. The use of such a pharmacokinetic/pharmacodynamic model defined a therapeutic window with an appropriate safety profile that enabled the clinical investigation of galunisertib. These efforts resulted in an intermittent dosing regimen (14 days on/14 days off, on a 28-day cycle) of galunisertib for all ongoing trials. Galunisertib is being investigated either as monotherapy or in combination with standard antitumor regimens (including nivolumab) in patients with cancer with high unmet medical needs such as glioblastoma, pancreatic cancer, and hepatocellular carcinoma. The present review summarizes the past and current experiences with different pharmacological treatments that enabled galunisertib to be investigated in patients.
Collapse
Affiliation(s)
| | - J Scott Sawyer
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Anja J Stauber
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Kyla E Driscoll
- Lilly Research Laboratories, Eli Lilly and Company, New York, NY, USA
| | - Shawn T Estrem
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Ann L Cleverly
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Durisala Desaiah
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Susan C Guba
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Karim A Benhadji
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Michael M Lahn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
50
|
Dubrulle J, Jordan BM, Akhmetova L, Farrell JA, Kim SH, Solnica-Krezel L, Schier AF. Response to Nodal morphogen gradient is determined by the kinetics of target gene induction. eLife 2015; 4. [PMID: 25869585 PMCID: PMC4395910 DOI: 10.7554/elife.05042] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 03/02/2015] [Indexed: 12/24/2022] Open
Abstract
Morphogen gradients expose cells to different signal concentrations and induce target genes with different ranges of expression. To determine how the Nodal morphogen gradient induces distinct gene expression patterns during zebrafish embryogenesis, we measured the activation dynamics of the signal transducer Smad2 and the expression kinetics of long- and short-range target genes. We found that threshold models based on ligand concentration are insufficient to predict the response of target genes. Instead, morphogen interpretation is shaped by the kinetics of target gene induction: the higher the rate of transcription and the earlier the onset of induction, the greater the spatial range of expression. Thus, the timing and magnitude of target gene expression can be used to modulate the range of expression and diversify the response to morphogen gradients. DOI:http://dx.doi.org/10.7554/eLife.05042.001 How a cell can tell where it is in a developing embryo has fascinated scientists for decades. The pioneering computer scientist and mathematical biologist Alan Turing was the first person to coin the term ‘morphogen’ to describe a protein that provides information about locations in the body. A morphogen is released from a group of cells (called the ‘source’) and as it moves away its activity (called the ‘signal’) declines gradually. Cells sense this signal gradient and use it to detect their position with respect to the source. Nodal is an important morphogen and is required to establish the correct identity of cells in the embryo; for example, it helps determine which cells should become a brain or heart or gut cell and so on. The zebrafish is a widely used model to study animal development, in part because its embryos are transparent; this allows cells and proteins to be easily observed under a microscope. When Nodal acts on cells, another protein called Smad2 becomes activated, moves into the cell's nucleus, and then binds to specific genes. This triggers the expression of these genes, which are first copied into mRNA molecules via a process known as transcription and are then translated into proteins. The protein products of these targeted genes control cell identity and movement. Several models have been proposed to explain how different concentrations of Nodal switch on the expression of different target genes; that is to say, to explain how a cell interprets the Nodal gradient. Dubrulle et al. have now measured factors that underlie how this gradient is interpreted. Individual cells in zebrafish embryos were tracked under a microscope, and Smad2 activation and gene expression were assessed. Dubrulle et al. found that, in contradiction to previous models, the amount of Nodal present on its own was insufficient to predict the target gene response. Instead, their analysis suggests that the size of each target gene's response depends on its rate of transcription and how quickly it is first expressed in response to Nodal. These findings of Dubrulle et al. suggest that timing and transcription rate are important in determining the appropriate response to Nodal. Further work will be now needed to find out whether similar mechanisms regulate other processes that rely on the activity of morphogens. DOI:http://dx.doi.org/10.7554/eLife.05042.002
Collapse
Affiliation(s)
- Julien Dubrulle
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Benjamin M Jordan
- Department of Mathematics, College of Science and Engineering, University of Minnesota, Minneapolis, United States
| | - Laila Akhmetova
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Jeffrey A Farrell
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Seok-Hyung Kim
- Division of Medicine, Medical University of South Carolina, Charleston, United States
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, United States
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| |
Collapse
|