Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters

Publication year range
1.
Nature ; 609(7929): 975-985, 2022 09.
Article in English | MEDLINE | ID: mdl-36104561

ABSTRACT

Understanding cell state transitions and purposefully controlling them is a longstanding challenge in biology. Here we present cell state transition assessment and regulation (cSTAR), an approach for mapping cell states, modelling transitions between them and predicting targeted interventions to convert cell fate decisions. cSTAR uses omics data as input, classifies cell states, and develops a workflow that transforms the input data into mechanistic models that identify a core signalling network, which controls cell fate transitions by influencing whole-cell networks. By integrating signalling and phenotypic data, cSTAR models how cells manoeuvre in Waddington's landscape1 and make decisions about which cell fate to adopt. Notably, cSTAR devises interventions to control the movement of cells in Waddington's landscape. Testing cSTAR in a cellular model of differentiation and proliferation shows a high correlation between quantitative predictions and experimental data. Applying cSTAR to different types of perturbation and omics datasets, including single-cell data, demonstrates its flexibility and scalability and provides new biological insights. The ability of cSTAR to identify targeted perturbations that interconvert cell fates will enable designer approaches for manipulating cellular development pathways and mechanistically underpinned therapeutic interventions.


Subject(s)
Cell Differentiation , Models, Biological , Signal Transduction , Cell Proliferation , Datasets as Topic , Phenotype , Single-Cell Analysis , Workflow
2.
Cell ; 141(5): 884-96, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20493519

ABSTRACT

Activation of ErbB receptors by epidermal growth factor (EGF) or heregulin (HRG) determines distinct cell-fate decisions, although signals propagate through shared pathways. Using mathematical modeling and experimental approaches, we unravel how HRG and EGF generate distinct, all-or-none responses of the phosphorylated transcription factor c-Fos. In the cytosol, EGF induces transient and HRG induces sustained ERK activation. In the nucleus, however, ERK activity and c-fos mRNA expression are transient for both ligands. Knockdown of dual-specificity phosphatases extends HRG-stimulated nuclear ERK activation, but not c-fos mRNA expression, implying the existence of a HRG-induced repressor of c-fos transcription. Further experiments confirmed that this repressor is mainly induced by HRG, but not EGF, and requires new protein synthesis. We show how a spatially distributed, signaling-transcription cascade robustly discriminates between transient and sustained ERK activities at the c-Fos system level. The proposed control mechanisms are general and operate in different cell types, stimulated by various ligands.


Subject(s)
Models, Biological , Proto-Oncogene Proteins c-fos/genetics , Cell Line, Tumor , Dual-Specificity Phosphatases/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Neuregulin-1/metabolism , Protein Stability , Proto-Oncogene Proteins c-fos/metabolism , Transcription, Genetic
3.
Cell ; 133(4): 566-7, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18485861

ABSTRACT

Spatial separation of component enzymes in a pathway creates microdomains and gradients of signaling activities. Imaging techniques and computational modeling help us to understand the complex relationships between signaling and spatial information. In this issue, Neves et al. (2008) report how the propagation of spatial information is controlled by the shape of hippocampal neurons and feedback and feed-forward motifs of the beta-adrenergic receptor signaling pathway.


Subject(s)
Neurons/cytology , Neurons/metabolism , Signal Transduction , Animals , Cell Shape , Feedback, Physiological , Hippocampus/cytology , Receptors, Adrenergic, beta-2/metabolism
4.
Nat Rev Mol Cell Biol ; 11(6): 414-26, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20495582

ABSTRACT

Although we have amassed extensive catalogues of signalling network components, our understanding of the spatiotemporal control of emergent network structures has lagged behind. Dynamic behaviour is starting to be explored throughout the genome, but analysis of spatial behaviours is still confined to individual proteins. The challenge is to reveal how cells integrate temporal and spatial information to determine specific biological functions. Key findings are the discovery of molecular signalling machines such as Ras nanoclusters, spatial activity gradients and flexible network circuitries that involve transcriptional feedback. They reveal design principles of spatiotemporal organization that are crucial for network function and cell fate decisions.


Subject(s)
Cell Communication , Signal Transduction , Animals , Cell Lineage , Humans , Time Factors , Transcription, Genetic
5.
Semin Cancer Biol ; 54: 162-173, 2019 02.
Article in English | MEDLINE | ID: mdl-29518522

ABSTRACT

RAS is the most frequently mutated gene across human cancers, but developing inhibitors of mutant RAS has proven to be challenging. Given the difficulties of targeting RAS directly, drugs that impact the other components of pathways where mutant RAS operates may potentially be effective. However, the system-level features, including different localizations of RAS isoforms, competition between downstream effectors, and interlocking feedback and feed-forward loops, must be understood to fully grasp the opportunities and limitations of inhibiting specific targets. Mathematical modeling can help us discern the system-level impacts of these features in normal and cancer cells. New technologies enable the acquisition of experimental data that will facilitate development of realistic models of oncogenic RAS behavior. In light of the wealth of empirical data accumulated over decades of study and the advancement of experimental methods for gathering new data, modelers now have the opportunity to advance progress toward realization of targeted treatment for mutant RAS-driven cancers.


Subject(s)
Gene Expression Regulation , Models, Biological , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism , Animals , Carrier Proteins , Drug Discovery , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , Protein Transport , Systems Biology/methods , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry
6.
Small ; 16(21): e1907476, 2020 05.
Article in English | MEDLINE | ID: mdl-32227434

ABSTRACT

Inhaled nanoparticles constitute a potential health hazard due to their size-dependent lung deposition and large surface to mass ratio. Exposure to high levels contributes to the risk of developing respiratory and cardiovascular diseases, as well as of lung cancer. Particle-induced acute phase response may be an important mechanism of action of particle-induced cardiovascular disease. Here, the authors review new important scientific evidence showing causal relationships between inhalation of particle and nanomaterials, induction of acute phase response, and risk of cardiovascular disease. Particle-induced acute phase response provides a means for risk assessment of particle-induced cardiovascular disease and underscores cardiovascular disease as an occupational disease.


Subject(s)
Acute-Phase Reaction , Cardiovascular Diseases , Inhalation Exposure , Nanoparticles , Acute-Phase Reaction/chemically induced , Cardiovascular Diseases/chemically induced , Humans , Inhalation Exposure/adverse effects , Lung/drug effects , Nanoparticles/toxicity , Occupational Diseases/chemically induced , Particle Size , Particulate Matter/toxicity
7.
PLoS Comput Biol ; 15(1): e1006706, 2019 01.
Article in English | MEDLINE | ID: mdl-30653502

ABSTRACT

Receptor tyrosine kinases (RTKs) typically contain multiple autophosphorylation sites in their cytoplasmic domains. Once activated, these autophosphorylation sites can recruit downstream signaling proteins containing Src homology 2 (SH2) and phosphotyrosine-binding (PTB) domains, which recognize phosphotyrosine-containing short linear motifs (SLiMs). These domains and SLiMs have polyspecific or promiscuous binding activities. Thus, multiple signaling proteins may compete for binding to a common SLiM and vice versa. To investigate the effects of competition on RTK signaling, we used a rule-based modeling approach to develop and analyze models for ligand-induced recruitment of SH2/PTB domain-containing proteins to autophosphorylation sites in the insulin-like growth factor 1 (IGF1) receptor (IGF1R). Models were parameterized using published datasets reporting protein copy numbers and site-specific binding affinities. Simulations were facilitated by a novel application of model restructuration, to reduce redundancy in rule-derived equations. We compare predictions obtained via numerical simulation of the model to those obtained through simple prediction methods, such as through an analytical approximation, or ranking by copy number and/or KD value, and find that the simple methods are unable to recapitulate the predictions of numerical simulations. We created 45 cell line-specific models that demonstrate how early events in IGF1R signaling depend on the protein abundance profile of a cell. Simulations, facilitated by model restructuration, identified pairs of IGF1R binding partners that are recruited in anti-correlated and correlated fashions, despite no inclusion of cooperativity in our models. This work shows that the outcome of competition depends on the physicochemical parameters that characterize pairwise interactions, as well as network properties, including network connectivity and the relative abundances of competitors.


Subject(s)
Models, Biological , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Animals , Binding Sites , Cell Line , Cluster Analysis , Computational Biology , Humans , Mice , Phosphorylation , Protein Binding , Proteins/chemistry , Proteins/metabolism , src Homology Domains
8.
J Biol Chem ; 293(6): 2206-2218, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29233889

ABSTRACT

Feedback control is a key mechanism in signal transduction, intimately involved in regulating the outcome of the cellular response. Here, we report a novel mechanism by which PHLDA1, Pleckstrin homology-like domain, family A, member 1, negatively regulates ErbB receptor signaling by inhibition of receptor oligomerization. We have found that the ErbB3 ligand, heregulin, induces PHILDA1 expression in MCF-7 cells. Transcriptionally-induced PHLDA1 protein directly binds to ErbB3, whereas knockdown of PHLDA1 increases complex formation between ErbB3 and ErbB2. To provide insight into the mechanism for our time-course and single-cell experimental observations, we performed a systematic computational search of network topologies of the mathematical models based on receptor dimer-tetramer formation in the ErbB activation processes. Our results indicate that only a model in which PHLDA1 inhibits formation of both dimers and tetramer can explain the experimental data. Predictions made from this model were further validated by single-molecule imaging experiments. Our studies suggest a unique regulatory feature of PHLDA1 to inhibit the ErbB receptor oligomerization process and thereby control the activity of receptor signaling network.


Subject(s)
Receptor, ErbB-3/metabolism , Transcription Factors/metabolism , Humans , MCF-7 Cells , Models, Chemical , Neuregulin-1/metabolism , Protein Multimerization , Signal Transduction , Single Molecule Imaging , Single-Cell Analysis , Transcription Factors/physiology , Transcription, Genetic
9.
Semin Cell Dev Biol ; 50: 85-94, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26481970

ABSTRACT

The notion of feedback is fundamental for understanding signal transduction networks. Feedback loops attenuate or amplify signals, change the network dynamics and modify the input-output relationships between the signal and the target. Negative feedback provides robustness to noise and adaptation to perturbations, but as a double-edged sword can prevent effective pathway inhibition by a drug. Positive feedback brings about switch-like network responses and can convert analog input signals into digital outputs, triggering cell fate decisions and phenotypic changes. We show how a multitude of protein-protein interactions creates hidden feedback loops in signal transduction cascades. Drug treatments that interfere with feedback regulation can cause unexpected adverse effects. Combinatorial molecular interactions generated by pathway crosstalk and feedback loops often bypass the block caused by targeted therapies against oncogenic mutated kinases. We discuss mechanisms of drug resistance caused by network adaptations and suggest that development of effective drug combinations requires understanding of how feedback loops modulate drug responses.


Subject(s)
Antineoplastic Agents/therapeutic use , Feedback, Physiological , Neoplasms/drug therapy , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Feedback, Physiological/drug effects , Homeostasis/drug effects , Humans , Signal Transduction/drug effects
10.
Semin Cell Dev Biol ; 58: 96-107, 2016 10.
Article in English | MEDLINE | ID: mdl-27350026

ABSTRACT

The intricate dynamic control and plasticity of RAS to ERK mitogenic, survival and apoptotic signalling has mystified researches for more than 30 years. Therapeutics targeting the oncogenic aberrations within this pathway often yield unsatisfactory, even undesired results, as in the case of paradoxical ERK activation in response to RAF inhibition. A direct approach of inhibiting single oncogenic proteins misses the dynamic network context governing the network signal processing. In this review, we discuss the signalling behaviour of RAS and RAF proteins in normal and in cancer cells, and the emerging systems-level properties of the RAS-to-ERK signalling network. We argue that to understand the dynamic complexities of this control system, mathematical models including mechanistic detail are required. Looking into the future, these dynamic models will build the foundation upon which more effective, rational approaches to cancer therapy will be developed.


Subject(s)
MAP Kinase Signaling System , Neoplasms/metabolism , ras Proteins/metabolism , Animals , Cell Lineage , Feedback, Physiological , Humans , Neoplasms/pathology
11.
Brief Bioinform ; 17(4): 593-602, 2016 07.
Article in English | MEDLINE | ID: mdl-26443615

ABSTRACT

The detailed, atomistic-level understanding of molecular signaling along the tumor-suppressive Hippo signaling pathway that controls tissue homeostasis by balancing cell proliferation and death through apoptosis is a promising avenue for the discovery of novel anticancer drug targets. The activation of kinases such as Mammalian STE20-Like Protein Kinases 1 and 2 (MST1 and MST2)-modulated through both homo- and heterodimerization (e.g. interactions with Ras association domain family, RASSF, enzymes)-is a key upstream event in this pathway and remains poorly understood. On the other hand, RASSFs (such as RASSF1A or RASSF5) act as important apoptosis activators and tumor suppressors, although their exact regulatory roles are also unclear. We present recent molecular studies of signaling along the Ras-RASSF-MST pathway, which controls growth and apoptosis in eukaryotic cells, including a variety of modern molecular modeling and simulation techniques. Using recently available structural information, we discuss the complex regulatory scenario according to which RASSFs perform dual signaling functions, either preventing or promoting MST2 activation, and thus control cell apoptosis. Here, we focus on recent studies highlighting the special role being played by the specific interactions between the helical Salvador/RASSF/Hippo (SARAH) domains of MST2 and RASSF1a or RASSF5 enzymes. These studies are crucial for integrating atomistic-level mechanistic information about the structures and conformational dynamics of interacting proteins, with information available on their system-level functions in cellular signaling.


Subject(s)
Protein Binding , Animals , Apoptosis , Models, Molecular , Protein Serine-Threonine Kinases , Signal Transduction
12.
PLoS Comput Biol ; 12(10): e1005051, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27716844

ABSTRACT

RASSF enzymes act as key apoptosis activators and tumor suppressors, being downregulated in many human cancers, although their exact regulatory roles remain unknown. A key downstream event in the RASSF pathway is the regulation of MST kinases, which are main effectors of RASSF-induced apoptosis. The regulation of MST1/2 includes both homo- and heterodimerization, mediated by helical SARAH domains, though the underlying molecular interaction mechanism is unclear. Here, we study the interactions between RASSF1A, RASSF5, and MST2 SARAH domains by using both atomistic molecular simulation techniques and experiments. We construct and study models of MST2 homodimers and MST2-RASSF SARAH heterodimers, and we identify the factors that control their high molecular stability. In addition, we also analyze both computationally and experimentally the interactions of MST2 SARAH domains with a series of synthetic peptides particularly designed to bind to it, and hope that our approach can be used to address some of the challenging problems in designing new anti-cancer drugs.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/ultrastructure , Cyclin-Dependent Kinase Inhibitor p15/chemistry , Cyclin-Dependent Kinase Inhibitor p15/ultrastructure , Drosophila Proteins/chemistry , Drosophila Proteins/ultrastructure , Molecular Docking Simulation , Binding Sites , Dimerization , Enzyme Activation , Protein Binding , Protein Conformation , Protein Domains
13.
Mol Syst Biol ; 11(11): 838, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26613961

ABSTRACT

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


Subject(s)
Cell Differentiation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Systems Biology/methods , Animals , Biosensing Techniques , Fluorescence Resonance Energy Transfer , Microfluidic Analytical Techniques , Models, Biological , PC12 Cells , Rats , Signal Transduction/drug effects
14.
Cell Mol Life Sci ; 72(12): 2431-43, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25697863

ABSTRACT

Cyclooxygenase 2 (COX2), a key regulatory enzyme of the prostaglandin/eicosanoid pathway, is an important target for anti-inflammatory therapy. It is highly induced by pro-inflammatory cytokines in a Nuclear factor kappa B (NFκB)-dependent manner. However, the mechanisms determining the amplitude and dynamics of this important pro-inflammatory event are poorly understood. Furthermore, there is significant difference between human and mouse COX2 expression in response to the inflammatory stimulus tumor necrosis factor alpha (TNFα). Here, we report the presence of a molecular logic AND gate composed of two NFκB response elements (NREs) which controls the expression of human COX2 in a switch-like manner. Combining quantitative kinetic modeling and thermostatistical analysis followed by experimental validation in iterative cycles, we show that the human COX2 expression machinery regulated by NFκB displays features of a logic AND gate. We propose that this provides a digital, noise-filtering mechanism for a tighter control of expression in response to TNFα, such that a threshold level of NFκB activation is required before the promoter becomes active and initiates transcription. This NFκB-regulated AND gate is absent in the mouse COX2 promoter, most likely contributing to its differential graded response in promoter activity and protein expression to TNFα. Our data suggest that the NFκB-regulated AND gate acts as a novel mechanism for controlling the expression of human COX2 to TNFα, and its absence in the mouse COX2 provides the foundation for further studies on understanding species-specific differential gene regulation.


Subject(s)
Cyclooxygenase 2/genetics , Gene Expression Regulation , Models, Theoretical , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Response Elements/genetics , Animals , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , HT29 Cells , Humans , Mice , NF-kappa B/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
15.
Proc Natl Acad Sci U S A ; 110(31): E2934-43, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23847209

ABSTRACT

Serine/threonine/tyrosine-interacting protein (STYX) is a catalytically inactive member of the dual-specificity phosphatases (DUSPs) family. Whereas the role of DUSPs in cellular signaling is well explored, the function of STYX is still unknown. Here, we identify STYX as a spatial regulator of ERK signaling. We used predictive-model simulation to test several hypotheses for possible modes of STYX action. We show that STYX localizes to the nucleus, competes with nuclear DUSP4 for binding to ERK, and acts as a nuclear anchor that regulates ERK nuclear export. Depletion of STYX increases ERK activity in both cytosol and nucleus. Importantly, depletion of STYX causes an ERK-dependent fragmentation of the Golgi apparatus and inhibits Golgi polarization and directional cell migration. Finally, we show that overexpression of STYX reduces ERK1/2 activation, thereby blocking PC12 cell differentiation. Overall, our results identify STYX as an important regulator of ERK1/2 signaling critical for cell migration and PC12 cell differentiation.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nuclear Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Enzyme Activation/genetics , Gene Knockdown Techniques , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Nuclear Proteins/genetics , PC12 Cells , Rats
16.
Angew Chem Int Ed Engl ; 55(3): 983-6, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26644280

ABSTRACT

RAF kinases are key players in the MAPK signaling pathway and are important targets for personalized cancer therapy. RAF dimerization is part of the physiological activation mechanism, together with phosphorylation, and is known to convey resistance to RAF inhibitors. Herein, molecular dynamics simulations are used to show that phosphorylation of a key N-terminal acidic (NtA) motif facilitates RAF dimerization by introducing several interprotomer salt bridges between the αC-helix and charged residues upstream of the NtA motif. Additionally, we show that the R-spine of RAF interacts with a conserved Trp residue in the vicinity of the NtA motif, connecting the active sites of two protomers and thereby modulating the cooperative interactions in the RAF dimer. Our findings provide a first structure-based mechanism for the auto-transactivation of RAF and could be generally applicable to other kinases, opening new pathways for overcoming dimerization-related drug resistance.


Subject(s)
Transcriptional Activation , raf Kinases/metabolism , Dimerization , Phosphorylation , Protein Conformation , raf Kinases/chemistry
17.
J Cell Sci ; 126(Pt 6): 1454-63, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23390316

ABSTRACT

Activation of the hypoxia-inducible factor (HIF) pathway is a critical step in the transcriptional response to hypoxia. Although many of the key proteins involved have been characterised, the dynamics of their interactions in generating this response remain unclear. In the present study, we have generated a comprehensive mathematical model of the HIF-1α pathway based on core validated components and dynamic experimental data, and confirm the previously described connections within the predicted network topology. Our model confirms previous work demonstrating that the steps leading to optimal HIF-1α transcriptional activity require sequential inhibition of both prolyl- and asparaginyl-hydroxylases. We predict from our model (and confirm experimentally) that there is residual activity of the asparaginyl-hydroxylase FIH (factor inhibiting HIF) at low oxygen tension. Furthermore, silencing FIH under conditions where prolyl-hydroxylases are inhibited results in increased HIF-1α transcriptional activity, but paradoxically decreases HIF-1α stability. Using a core module of the HIF network and mathematical proof supported by experimental data, we propose that asparaginyl hydroxylation confers a degree of resistance upon HIF-1α to proteosomal degradation. Thus, through in vitro experimental data and in silico predictions, we provide a comprehensive model of the dynamic regulation of HIF-1α transcriptional activity by hydroxylases and use its predictive and adaptive properties to explain counter-intuitive biological observations.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mixed Function Oxygenases/metabolism , Models, Biological , Repressor Proteins/metabolism , Computational Biology , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/pharmacology , Oxygen/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Repressor Proteins/pharmacology , Signal Transduction , Transcriptional Activation/genetics
18.
PLoS Comput Biol ; 10(2): e1003385, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24550716

ABSTRACT

The ever-increasing capacity of biological molecular data acquisition outpaces our ability to understand the meaningful relationships between molecules in a cell. Multiple databases were developed to store and organize these molecular data. However, emerging fundamental questions about concerted functions of these molecules in hierarchical cellular networks are poorly addressed. Here we review recent advances in the development of publically available databases that help us analyze the signal integration and processing by multilayered networks that specify biological responses in model organisms and human cells.


Subject(s)
Databases, Factual , Models, Biological , Signal Transduction , Animals , Computational Biology , Drug Discovery , Humans , Systems Integration
19.
Cancers (Basel) ; 16(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39001416

ABSTRACT

Understanding signaling patterns of transformation and controlling cell phenotypes is a challenge of current biology. Here we applied a cell State Transition Assessment and Regulation (cSTAR) approach to a perturbation dataset of single cell phosphoproteomic patterns of multiple breast cancer (BC) and normal breast tissue-derived cell lines. Following a separation of luminal, basal, and normal cell states, we identified signaling nodes within core control networks, delineated causal connections, and determined the primary drivers underlying oncogenic transformation and transitions across distinct BC subtypes. Whereas cell lines within the same BC subtype have different mutational and expression profiles, the architecture of the core network was similar for all luminal BC cells, and mTOR was a main oncogenic driver. In contrast, core networks of basal BC were heterogeneous and segregated into roughly four major subclasses with distinct oncogenic and BC subtype drivers. Likewise, normal breast tissue cells were separated into two different subclasses. Based on the data and quantified network topologies, we derived mechanistic cSTAR models that serve as digital cell twins and allow the deliberate control of cell movements within a Waddington landscape across different cell states. These cSTAR models suggested strategies of normalizing phosphorylation networks of BC cell lines using small molecule inhibitors.

20.
bioRxiv ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38328237

ABSTRACT

A key feature of arteriogenesis is capillary-to-arterial endothelial cell fate transition. Although a number of studies in the past two decades suggested this process is driven by VEGF activation of Notch signaling, how arteriogenesis is regulated remains poorly understood. Here we report that arterial specification is mediated by fluid shear stress (FSS) independent of VEGFR2 signaling and that a decline in VEGFR2 signaling is required for arteriogenesis to fully take place. VEGF does not induce arterial fate in capillary ECs and, instead, counteracts FSS-driven capillary-to-arterial cell fate transition. Mechanistically, FSS-driven arterial program involves both Notch-dependent and Notch-independent events. Sox17 is the key mediator of the FSS-induced arterial specification and a target of VEGF-FSS competition. These findings suggest a new paradigm of VEGF-FSS crosstalk coordinating angiogenesis, arteriogenesis and capillary maintenance.

SELECTION OF CITATIONS
SEARCH DETAIL