Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(9): 1499-1510, 2023 09.
Article in English | MEDLINE | ID: mdl-37500885

ABSTRACT

Chimeric antigen receptor (CAR)-T cells are powerful therapeutics; however, their efficacy is often hindered by critical hurdles. Here utilizing the endocytic feature of the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) cytoplasmic tail, we reprogram CAR function and substantially enhance CAR-T efficacy in vivo. CAR-T cells with monomeric, duplex or triplex CTLA-4 cytoplasmic tails (CCTs) fused to the C terminus of CAR exhibit a progressive increase in cytotoxicity under repeated stimulation, accompanied by reduced activation and production of proinflammatory cytokines. Further characterization reveals that CARs with increasing CCT fusion show a progressively lower surface expression, regulated by their constant endocytosis, recycling and degradation under steady state. The molecular dynamics of reengineered CAR with CCT fusion results in reduced CAR-mediated trogocytosis, loss of tumor antigen and improved CAR-T survival. CARs with either monomeric (CAR-1CCT) or duplex CCTs (CAR-2CCT) have superior antitumor efficacy in a relapsed leukemia model. Single-cell RNA sequencing and flow cytometry analysis reveal that CAR-2CCT cells retain a stronger central memory phenotype and exhibit increased persistence. These findings illuminate a unique strategy for engineering therapeutic T cells and improving CAR-T function through synthetic CCT fusion, which is orthogonal to other cell engineering techniques.


Subject(s)
Receptors, Chimeric Antigen , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , CTLA-4 Antigen/genetics , Immunotherapy, Adoptive/methods , T-Lymphocytes , Cytokines/metabolism , Abatacept , Receptors, Antigen, T-Cell/genetics , Cell Line, Tumor
2.
Mol Cell ; 81(4): 708-723.e5, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606974

ABSTRACT

The PI3K pathway regulates cell metabolism, proliferation, and migration, and its dysregulation is common in cancer. We now show that both physiologic and oncogenic activation of PI3K signaling increase the expression of its negative regulator PTEN. This limits the duration of the signal and output of the pathway. Physiologic and pharmacologic inhibition of the pathway reduces PTEN and contributes to the rebound in pathway activity in tumors treated with PI3K inhibitors and limits their efficacy. Regulation of PTEN is due to mTOR/4E-BP1-dependent control of its translation and is lost when 4E-BP1 is deleted. Translational regulation of PTEN is therefore a major homeostatic regulator of physiologic PI3K signaling and plays a role in reducing the pathway activation by oncogenic PIK3CA mutants and the antitumor activity of PI3K pathway inhibitors. However, pathway output is hyperactivated in tumor cells with coexistent PI3K mutation and loss of PTEN function.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Homeostasis , Neoplasms/enzymology , PTEN Phosphohydrolase/biosynthesis , Protein Biosynthesis , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CHO Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Class I Phosphatidylinositol 3-Kinases/genetics , Cricetulus , Humans , Mutation , Neoplasms/genetics , PTEN Phosphohydrolase/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
Mol Cell ; 72(1): 60-70.e3, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30244832

ABSTRACT

Epigenetic control of regulatory networks is only partially understood. Expression of Insulin-like growth factor-II (IGF2) is controlled by genomic imprinting, mediated by silencing of the maternal allele. Loss of imprinting of IGF2 (LOI) is linked to intestinal and colorectal cancers, causally in murine models and epidemiologically in humans. However, the molecular underpinnings of the LOI phenotype are not clear. Surprisingly, in LOI cells, we find a reversal of the relative activities of two canonical signaling pathways triggered by IGF2, causing further rebalancing between pro- and anti-apoptotic signaling. A predictive mathematical model shows that this network rebalancing quantitatively accounts for the effect of receptor tyrosine kinase inhibition in both WT and LOI cells. This mechanism also quantitatively explains both the stable LOI phenotype and the therapeutic window for selective killing of LOI cells, and thus prevention of epigenetically controlled cancers. These findings suggest a framework for understanding epigenetically modified cell signaling.


Subject(s)
Colorectal Neoplasms/genetics , Epigenesis, Genetic/genetics , Genomic Imprinting/genetics , Insulin-Like Growth Factor II/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Mice , Phenotype , Signal Transduction
4.
Am J Pathol ; 194(7): 1171-1184, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38548268

ABSTRACT

Interactions between endothelial cells (ECs) and mural pericytes (PCs) are critical in maintaining the stability and function of the microvascular wall. Abnormal interactions between these two cell types are a hallmark of progressive fibrotic diseases such as systemic sclerosis (also known as scleroderma). However, the role of PCs in signaling microvascular dysfunction remains underexplored. We hypothesized that integrin-matrix interactions contribute to PC migration from the vascular wall and conversion into interstitial myofibroblasts. Herein, pro-inflammatory tumor necrosis factor α (TNFα) or a fibrotic growth factor [transforming growth factor ß1 (TGF-ß1)] were used to evaluate human PC inflammatory and fibrotic phenotypes by assessing their migration, matrix deposition, integrin expression, and subsequent effects on endothelial dysfunction. Both TNFα and TGF-ß1 treatment altered integrin expression and matrix protein deposition, but only fibrotic TGF-ß1 drove PC migration in an integrin-dependent manner. In addition, integrin-dependent PC migration was correlated to changes in EC angiopoietin-2 levels, a marker of vascular instability. Finally, there was evidence of changes in vascular stability corresponding to disease state in human systemic sclerosis skin. This work shows that TNFα and TGF-ß1 induce changes in PC integrin expression and matrix deposition that facilitate migration and reduce vascular stability, providing evidence that microvascular destabilization can be an early indicator of tissue fibrosis.


Subject(s)
Cell Movement , Fibrosis , Integrins , Pericytes , Scleroderma, Systemic , Transforming Growth Factor beta1 , Pericytes/metabolism , Pericytes/pathology , Humans , Transforming Growth Factor beta1/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/metabolism , Integrins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Microvessels/pathology , Microvessels/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Skin/pathology , Skin/metabolism , Skin/blood supply
5.
Mol Cell Neurosci ; 123: 103772, 2022 12.
Article in English | MEDLINE | ID: mdl-36055521

ABSTRACT

Axon guidance during neural wiring involves a series of precisely controlled chemotactic events by the motile axonal tip, the growth cone. A fundamental question is how neuronal growth cones make directional decisions in response to extremely shallow gradients of guidance cues with exquisite sensitivity. Here we report that nerve growth cones possess a signal amplification mechanism during gradient sensing process. In neuronal growth cones of Xenopus spinal neurons, phosphatidylinositol-3,4,5-trisphosphate (PIP3), an important signaling molecule in chemotaxis, was actively recruited to the up-gradient side in response to an external gradient of brain-derived neurotrophic factor (BDNF), resulting in an intracellular gradient with approximate 30-fold amplification of the input. Furthermore, a reverse gradient of phosphatase and tensin homolog (PTEN) was induced by BDNF within the growth cone and the increased PTEN activity at the down-gradient side is required for the amplification of PIP3 signals. Mechanistically, the establishment of both positive PIP3 and reverse PTEN gradients depends on the filamentous actin network. Together with computational modeling, our results revealed a double negative feedback loop among PTEN, PIP3 and actomyosin for signal amplification, which is essential for gradient sensing of neuronal growth cones in response to diffusible cues.


Subject(s)
Actomyosin , Growth Cones , Growth Cones/physiology , Brain-Derived Neurotrophic Factor , Feedback , Chemotaxis/physiology
6.
Proc Natl Acad Sci U S A ; 116(47): 23551-23561, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685607

ABSTRACT

Angiogenesis frequently occurs in the context of acute or persistent inflammation. The complex interplay of proinflammatory and proangiogenic cues is only partially understood. Using an experimental model, permitting exposure of developing blood vessel sprouts to multiple combinations of diverse biochemical stimuli and juxtacrine cell interactions, we present evidence that a proinflammatory cytokine, tumor necrosis factor (TNF), can have both proangiogenic and antiangiogenic effects, depending on the dose and the presence of pericytes. In particular, we find that pericytes can rescue and enhance angiogenesis in the presence of otherwise-inhibitory high TNF doses. This sharp switch from proangiogenic to antiangiogenic effect of TNF observed with an escalating dose of this cytokine, as well as the effect of pericytes, are explained by a mathematical model trained on the biochemical data. Furthermore, this model was predictive of the effects of diverse combinations of proinflammatory and antiinflammatory cues, and variable pericyte coverage. The mechanism supports the effect of TNF and pericytes as modulating signaling networks impinging on Notch signaling and specification of the Tip and Stalk phenotypes. This integrative analysis elucidates the plasticity of the angiogenic morphogenesis in the presence of diverse and potentially conflicting cues, with immediate implications for many physiological and pathological settings.


Subject(s)
Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Pericytes/physiology , Tumor Necrosis Factor-alpha/pharmacology , Cell Communication , Cell Culture Techniques , Coculture Techniques , Dose-Response Relationship, Drug , Fibroblast Growth Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Lysophospholipids/pharmacology , Models, Biological , Neovascularization, Pathologic/pathology , Pericytes/drug effects , Receptors, Notch/physiology , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tissue Engineering , Tumor Necrosis Factor-alpha/administration & dosage , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/physiology
7.
Proc Natl Acad Sci U S A ; 116(28): 14374-14383, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31239339

ABSTRACT

Transplanted stromal cells have demonstrated considerable promise as therapeutic agents in diverse disease settings. Paracrine signaling can be an important mediator of these therapeutic effects at the sites of acute or persistent injury and inflammation. As many stromal cell types, including bone marrow-derived stromal cells (BMSCs), display tissue-specific responses, there is a need to explore their secretory dynamics in the context of tissue and injury type. Paracrine signals are not static, and could encode contextual dynamics in the kinetic changes of the concentrations of the secreted ligands. However, precise measurement of dynamic and context-specific cellular secretory signatures, particularly in adherent cells, remains challenging. Here, by creating an experimental and computational analysis platform, we reconstructed dynamic secretory signatures of cells based on a very limited number of time points. By using this approach, we demonstrate that the secretory signatures of CD133-positive BMSCs are uniquely defined by distinct biological contexts, including signals from injured cardiac cells undergoing oxidative stress, characteristic of cardiac infarction. Furthermore, we show that the mixture of recombinant factors reproducing the dynamics of BMSC-generated secretion can mediate a highly effective rescue of cells injured by oxidative stress and an improved cardiac output. These results support the importance of the dynamic multifactorial paracrine signals in mediating remedial effects of stromal stem cells, and pave the way for stem cell-inspired cell-free treatments of cardiac and other injuries.


Subject(s)
Inflammation/genetics , Mesenchymal Stem Cells , Myocardial Infarction/genetics , Neovascularization, Physiologic/genetics , AC133 Antigen/genetics , Animals , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation/genetics , Cells, Cultured , Humans , Inflammation/metabolism , Inflammation/pathology , Ligands , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Oxidative Stress/genetics , Paracrine Communication/genetics
8.
Proc Natl Acad Sci U S A ; 114(21): 5503-5508, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28500274

ABSTRACT

Cerebral cavernous malformations (CCMs) are common vascular anomalies that develop in the central nervous system and, more rarely, the retina. The lesions can cause headache, seizures, focal neurological deficits, and hemorrhagic stroke. Symptomatic lesions are treated according to their presentation; however, targeted pharmacological therapies that improve the outcome of CCM disease are currently lacking. We performed a high-throughput screen to identify Food and Drug Administration-approved drugs or other bioactive compounds that could effectively suppress hyperproliferation of mouse brain primary astrocytes deficient for CCM3. We demonstrate that fluvastatin, an inhibitor of 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase and the N-bisphosphonate zoledronic acid monohydrate, an inhibitor of protein prenylation, act synergistically to reverse outcomes of CCM3 loss in cultured mouse primary astrocytes and in Drosophila glial cells in vivo. Further, the two drugs effectively attenuate neural and vascular deficits in chronic and acute mouse models of CCM3 loss in vivo, significantly reducing lesion burden and extending longevity. Sustained inhibition of the mevalonate pathway represents a potential pharmacological treatment option and suggests advantages of combination therapy for CCM disease.


Subject(s)
Diphosphonates/therapeutic use , Fatty Acids, Monounsaturated/therapeutic use , Hemangioma, Cavernous, Central Nervous System/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Imidazoles/therapeutic use , Indoles/therapeutic use , Animals , Astrocytes/drug effects , Diphosphonates/pharmacology , Drosophila , Drug Evaluation, Preclinical , Drug Therapy, Combination , Endothelial Cells/drug effects , Female , Fluvastatin , High-Throughput Screening Assays , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mice , Pregnancy , Protein Prenylation/drug effects , Zoledronic Acid
9.
Proc Natl Acad Sci U S A ; 114(28): E5750-E5759, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28655842

ABSTRACT

Cell polarization and directional cell migration can display random, persistent, and oscillatory dynamic patterns. However, it is not clear whether these polarity patterns can be explained by the same underlying regulatory mechanism. Here, we show that random, persistent, and oscillatory migration accompanied by polarization can simultaneously occur in populations of melanoma cells derived from tumors with different degrees of aggressiveness. We demonstrate that all of these patterns and the probabilities of their occurrence are quantitatively accounted for by a simple mechanism involving a spatially distributed, mechanochemical feedback coupling the dynamically changing extracellular matrix (ECM)-cell contacts to the activation of signaling downstream of the Rho-family small GTPases. This mechanism is supported by a predictive mathematical model and extensive experimental validation, and can explain previously reported results for diverse cell types. In melanoma, this mechanism also accounts for the effects of genetic and environmental perturbations, including mutations linked to invasive cell spread. The resulting mechanistic understanding of cell polarity quantitatively captures the relationship between population variability and phenotypic plasticity, with the potential to account for a wide variety of cell migration states in diverse pathological and physiological conditions.


Subject(s)
Cell Polarity/physiology , Feedback, Physiological , Melanoma/metabolism , Skin Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Shape , Disease Progression , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Melanoma/pathology , Models, Theoretical , Mutation , Neoplasm Invasiveness , Oscillometry , Phenotype , Signal Transduction , Skin Neoplasms/pathology , rho GTP-Binding Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 113(6): E689-95, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26792517

ABSTRACT

Gradient sensing requires at least two measurements at different points in space. These measurements must then be communicated to a common location to be compared, which is unavoidably noisy. Although much is known about the limits of measurement precision by cells, the limits placed by the communication are not understood. Motivated by recent experiments, we derive the fundamental limits to the precision of gradient sensing in a multicellular system, accounting for communication and temporal integration. The gradient is estimated by comparing a "local" and a "global" molecular reporter of the external concentration, where the global reporter is exchanged between neighboring cells. Using the fluctuation-dissipation framework, we find, in contrast to the case when communication is ignored, that precision saturates with the number of cells independently of the measurement time duration, because communication establishes a maximum length scale over which sensory information can be reliably conveyed. Surprisingly, we also find that precision is improved if the local reporter is exchanged between cells as well, albeit more slowly than the global reporter. The reason is that whereas exchange of the local reporter weakens the comparison, it decreases the measurement noise. We term such a model "regional excitation-global inhibition." Our results demonstrate that fundamental sensing limits are necessarily sharpened when the need to communicate information is taken into account.


Subject(s)
Cell Communication , Models, Biological , Signal-To-Noise Ratio , Time Factors
11.
Proc Natl Acad Sci U S A ; 113(40): E5896-E5905, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27651485

ABSTRACT

In response to pheromones, yeast cells activate a MAPK pathway to direct processes important for mating, including gene induction, cell-cycle arrest, and polarized cell growth. Although a variety of assays have been able to elucidate signaling activities at multiple steps in the pathway, measurements of MAPK activity during the pheromone response have remained elusive, and our understanding of single-cell signaling behavior is incomplete. Using a yeast-optimized FRET-based mammalian Erk-activity reporter to monitor Fus3 and Kss1 activity in live yeast cells, we demonstrate that overall mating MAPK activity exhibits distinct temporal dynamics, rapid reversibility, and a graded dose dependence around the KD of the receptor, where phenotypic transitions occur. The complex dose response was found to be largely a consequence of two feedbacks involving cyclin-mediated scaffold phosphorylation and Fus3 autoregulation. Distinct cell cycle-dependent response patterns comprised a large portion of the cell-to-cell variability at each dose, constituting the major source of extrinsic noise in coupling activity to downstream gene-expression responses. Additionally, we found diverse spatial MAPK activity patterns to emerge over time in cells undergoing default, gradient, and true mating responses. Furthermore, ramping up and rapid loss of activity were closely associated with zygote formation in mating-cell pairs, supporting a role for elevated MAPK activity in successful cell fusion and morphogenic reorganization. Altogether, these findings present a detailed view of spatiotemporal MAPK activity during the pheromone response, elucidating its role in mediating complex long-term developmental fates in a unicellular differentiation system.


Subject(s)
Cell Differentiation , Mitogen-Activated Protein Kinases/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Single-Cell Analysis/methods , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Fusion , Cell Polarity/drug effects , Enzyme Activation/drug effects , MAP Kinase Signaling System/drug effects , Pheromones/pharmacology , Phosphorylation/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Time-Lapse Imaging
12.
Proc Natl Acad Sci U S A ; 113(30): 8496-501, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27402752

ABSTRACT

Most strains of rhinovirus (RV), the common cold virus, replicate better at cool temperatures found in the nasal cavity (33-35 °C) than at lung temperature (37 °C). Recent studies found that although 37 °C temperature suppressed RV growth largely by engaging the type 1 IFN response in infected epithelial cells, a significant temperature dependence to viral replication remained in cells devoid of IFN induction or signaling. To gain insight into IFN-independent mechanisms limiting RV replication at 37 °C, we studied RV infection in human bronchial epithelial cells and H1-HeLa cells. During the single replication cycle, RV exhibited temperature-dependent replication in both cell types in the absence of IFN induction. At 37 °C, earlier signs of apoptosis in RV-infected cells were accompanied by reduced virus production. Furthermore, apoptosis of epithelial cells was enhanced at 37 °C in response to diverse stimuli. Dynamic mathematical modeling and B cell lymphoma 2 (BCL2) overexpression revealed that temperature-dependent host cell death could partially account for the temperature-dependent growth observed during RV amplification, but also suggested additional mechanisms of virus control. In search of a redundant antiviral pathway, we identified a role for the RNA-degrading enzyme RNAseL. Simultaneous antagonism of apoptosis and RNAseL increased viral replication and dramatically reduced temperature dependence. These findings reveal two IFN-independent mechanisms active in innate defense against RV, and demonstrate that even in the absence of IFNs, temperature-dependent RV amplification is largely a result of host cell antiviral restriction mechanisms operating more effectively at 37 °C than at 33 °C.


Subject(s)
Interferons/genetics , RNA, Double-Stranded/genetics , Rhinovirus/genetics , Temperature , Virus Replication/genetics , A549 Cells , Apoptosis/genetics , Bronchi/cytology , Cells, Cultured , Common Cold/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression , HeLa Cells , Humans , Interferons/metabolism , Respiratory Mucosa/cytology , Rhinovirus/physiology
13.
Proc Natl Acad Sci U S A ; 113(6): E679-88, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26792522

ABSTRACT

Collective cell responses to exogenous cues depend on cell-cell interactions. In principle, these can result in enhanced sensitivity to weak and noisy stimuli. However, this has not yet been shown experimentally, and little is known about how multicellular signal processing modulates single-cell sensitivity to extracellular signaling inputs, including those guiding complex changes in the tissue form and function. Here we explored whether cell-cell communication can enhance the ability of cell ensembles to sense and respond to weak gradients of chemotactic cues. Using a combination of experiments with mammary epithelial cells and mathematical modeling, we find that multicellular sensing enables detection of and response to shallow epidermal growth factor (EGF) gradients that are undetectable by single cells. However, the advantage of this type of gradient sensing is limited by the noisiness of the signaling relay, necessary to integrate spatially distributed ligand concentration information. We calculate the fundamental sensory limits imposed by this communication noise and combine them with the experimental data to estimate the effective size of multicellular sensory groups involved in gradient sensing. Functional experiments strongly implicated intercellular communication through gap junctions and calcium release from intracellular stores as mediators of collective gradient sensing. The resulting integrative analysis provides a framework for understanding the advantages and limitations of sensory information processing by relays of chemically coupled cells.


Subject(s)
Cell Communication , Morphogenesis , Animals , Cadherins/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cell Communication/drug effects , Cell Movement/drug effects , Computer Simulation , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Gap Junctions/drug effects , Gap Junctions/metabolism , Ions , Ligands , Mammary Glands, Animal/cytology , Models, Biological , Morphogenesis/drug effects , Organoids/cytology , Organoids/drug effects , Rats , Time Factors
14.
Biophys J ; 114(6): 1257-1263, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29590582

ABSTRACT

Living cells orient the cytoskeleton polarity and directional migration in response to spatial gradients of multiple types of cues. The resulting tactic behaviors are critical for the proper cell localization in the context of complex single-cell and tissue behaviors. In this perspective, we highlight the recent discovery of, to our knowledge, a new -taxis phenomenon, the topotaxis, which mediates directional cell migration in response to the gradients of such topographic features as the density of extracellular matrix fibers. The direction of topotactic migration critically depends on the effective stiffness of the cortical cytoskeleton, which is controlled by the balance between two parallel signaling pathways activated by the extracellular matrix input. Topotaxis can account for such striking cell behaviors as the opposite directionality of migration of benign and metastatic cancer cells and certain aspects of the wound-healing process. We anticipate that, in conjunction with other tactic phenomena, topotaxis can provide critical information for understanding and design of tissue structure and function.


Subject(s)
Cell Movement , Extracellular Matrix/metabolism , Biomechanical Phenomena , Humans , Neoplasm Metastasis , Neoplasms/pathology
15.
PLoS Comput Biol ; 13(5): e1005524, 2017 05.
Article in English | MEDLINE | ID: mdl-28472054

ABSTRACT

Protrusion and retraction of lamellipodia are common features of eukaryotic cell motility. As a cell migrates through its extracellular matrix (ECM), lamellipod growth increases cell-ECM contact area and enhances engagement of integrin receptors, locally amplifying ECM input to internal signaling cascades. In contrast, contraction of lamellipodia results in reduced integrin engagement that dampens the level of ECM-induced signaling. These changes in cell shape are both influenced by, and feed back onto ECM signaling. Motivated by experimental observations on melanoma cells lines (1205Lu and SBcl2) migrating on fibronectin (FN) coated topographic substrates (anisotropic post-density arrays), we probe this interplay between intracellular and ECM signaling. Experimentally, cells exhibited one of three lamellipodial dynamics: persistently polarized, random, or oscillatory, with competing lamellipodia oscillating out of phase (Park et al., 2017). Pharmacological treatments, changes in FN density, and substrate topography all affected the fraction of cells exhibiting these behaviours. We use these observations as constraints to test a sequence of hypotheses for how intracellular (GTPase) and ECM signaling jointly regulate lamellipodial dynamics. The models encoding these hypotheses are predicated on mutually antagonistic Rac-Rho signaling, Rac-mediated protrusion (via activation of Arp2/3 actin nucleation) and Rho-mediated contraction (via ROCK phosphorylation of myosin light chain), which are coupled to ECM signaling that is modulated by protrusion/contraction. By testing each model against experimental observations, we identify how the signaling layers interact to generate the diverse range of cell behaviors, and how various molecular perturbations and changes in ECM signaling modulate the fraction of cells exhibiting each. We identify several factors that play distinct but critical roles in generating the observed dynamic: (1) competition between lamellipodia for shared pools of Rac and Rho, (2) activation of RhoA by ECM signaling, and (3) feedback from lamellipodial growth or contraction to cell-ECM contact area and therefore to the ECM signaling level.


Subject(s)
Cell Physiological Phenomena/physiology , Models, Biological , Computational Biology , Extracellular Matrix/physiology , rho GTP-Binding Proteins/metabolism
16.
PLoS Comput Biol ; 13(4): e1005436, 2017 04.
Article in English | MEDLINE | ID: mdl-28379950

ABSTRACT

In this study a new computational method is developed to quantify decision making errors in cells, caused by noise and signaling failures. Analysis of tumor necrosis factor (TNF) signaling pathway which regulates the transcription factor Nuclear Factor κB (NF-κB) using this method identifies two types of incorrect cell decisions called false alarm and miss. These two events represent, respectively, declaring a signal which is not present and missing a signal that does exist. Using single cell experimental data and the developed method, we compute false alarm and miss error probabilities in wild-type cells and provide a formulation which shows how these metrics depend on the signal transduction noise level. We also show that in the presence of abnormalities in a cell, decision making processes can be significantly affected, compared to a wild-type cell, and the method is able to model and measure such effects. In the TNF-NF-κB pathway, the method computes and reveals changes in false alarm and miss probabilities in A20-deficient cells, caused by cell's inability to inhibit TNF-induced NF-κB response. In biological terms, a higher false alarm metric in this abnormal TNF signaling system indicates perceiving more cytokine signals which in fact do not exist at the system input, whereas a higher miss metric indicates that it is highly likely to miss signals that actually exist. Overall, this study demonstrates the ability of the developed method for modeling cell decision making errors under normal and abnormal conditions, and in the presence of transduction noise uncertainty. Compared to the previously reported pathway capacity metric, our results suggest that the introduced decision error metrics characterize signaling failures more accurately. This is mainly because while capacity is a useful metric to study information transmission in signaling pathways, it does not capture the overlap between TNF-induced noisy response curves.


Subject(s)
Cell Communication/physiology , Computational Biology/methods , Models, Biological , Models, Statistical , Signal Transduction/physiology , Decision Theory , NF-kappa B/metabolism , Signal Processing, Computer-Assisted , Single-Cell Analysis , Tumor Necrosis Factor-alpha/metabolism
17.
Nat Mater ; 15(7): 792-801, 2016 07.
Article in English | MEDLINE | ID: mdl-26974411

ABSTRACT

Living cells and the extracellular matrix (ECM) can exhibit complex interactions that define key developmental, physiological and pathological processes. Here, we report a new type of directed migration-which we term 'topotaxis'-guided by the gradient of the nanoscale topographic features in the cells' ECM environment. We show that the direction of topotaxis is reflective of the effective cell stiffness, and that it depends on the balance of the ECM-triggered signalling pathways PI(3)K-Akt and ROCK-MLCK. In melanoma cancer cells, this balance can be altered by different ECM inputs, pharmacological perturbations or genetic alterations, particularly a loss of PTEN in aggressive melanoma cells. We conclude that topotaxis is a product of the material properties of cells and the surrounding ECM, and propose that the invasive capacity of many cancers may depend broadly on topotactic responses, providing a potentially attractive mechanism for controlling invasive and metastatic behaviour.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic/physiology , Melanoma , Taxis Response/physiology , Cell Line, Tumor , Humans , Melanoma/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Surface Properties , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
18.
Proc Natl Acad Sci U S A ; 111(48): 17330-5, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404303

ABSTRACT

Experimental measurements of biochemical noise have primarily focused on sources of noise at the gene expression level due to limitations of existing noise decomposition techniques. Here, we introduce a mathematical framework that extends classical extrinsic-intrinsic noise analysis and enables mapping of noise within upstream signaling networks free of such restrictions. The framework applies to systems for which the responses of interest are linearly correlated on average, although the framework can be easily generalized to the nonlinear case. Interestingly, despite the high degree of complexity and nonlinearity of most mammalian signaling networks, three distinct tumor necrosis factor (TNF) signaling network branches displayed linearly correlated responses, in both wild-type and perturbed versions of the network, across multiple orders of magnitude of ligand concentration. Using the noise mapping analysis, we find that the c-Jun N-terminal kinase (JNK) pathway generates higher noise than the NF-κB pathway, whereas the activation of c-Jun adds a greater amount of noise than the activation of ATF-2. In addition, we find that the A20 protein can suppress noise in the activation of ATF-2 by separately inhibiting the TNF receptor complex and JNK pathway through a negative feedback mechanism. These results, easily scalable to larger and more complex networks, pave the way toward assessing how noise propagates through cellular signaling pathways and create a foundation on which we can further investigate the relationship between signaling system architecture and biological noise.


Subject(s)
Algorithms , Biochemical Phenomena/physiology , Intracellular Space/metabolism , Models, Biological , Signal Transduction/physiology , 3T3 Cells , Activating Transcription Factor 2/metabolism , Animals , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Feedback, Physiological/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Space/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Microscopy, Fluorescence , Mutation , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factors/pharmacology
19.
Phys Biol ; 13(3): 035004, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27203129

ABSTRACT

Gradient sensing underlies important biological processes including morphogenesis, polarization, and cell migration. The precision of gradient sensing increases with the length of a detector (a cell or group of cells) in the gradient direction, since a longer detector spans a larger range of concentration values. Intuition from studies of concentration sensing suggests that precision should also increase with detector length in the direction transverse to the gradient, since then spatial averaging should reduce the noise. However, here we show that, unlike for concentration sensing, the precision of gradient sensing decreases with transverse length for the simplest gradient sensing model, local excitation-global inhibition. The reason is that gradient sensing ultimately relies on a subtraction of measured concentration values. While spatial averaging indeed reduces the noise in these measurements, which increases precision, it also reduces the covariance between the measurements, which results in the net decrease in precision. We demonstrate how a recently introduced gradient sensing mechanism, regional excitation-global inhibition (REGI), overcomes this effect and recovers the benefit of transverse averaging. Using a REGI-based model, we compute the optimal two- and three-dimensional detector shapes, and argue that they are consistent with the shapes of naturally occurring gradient-sensing cell populations.


Subject(s)
Chemotaxis , Models, Biological , Signal Transduction , Cell Movement
20.
J Mol Cell Cardiol ; 86: 168-78, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26241846

ABSTRACT

cAMP-PKA protein kinase is a key nodal signaling pathway that regulates a wide range of heart pacemaker cell functions. These functions are predicted to be involved in regulation of spontaneous action potential (AP) generation of these cells. Here we investigate if the kinetics and stoichiometry of increase in PKA activity match the increase in AP firing rate in response to ß-adrenergic receptor (ß-AR) stimulation or phosphodiesterase (PDE) inhibition, that alters the AP firing rate of heart sinoatrial pacemaker cells. In cultured adult rabbit pacemaker cells infected with an adenovirus expressing the FRET sensor AKAR3, the EC50 in response to graded increases in the intensity of ß-AR stimulation (by Isoproterenol) the magnitude of the increases in PKA activity and the spontaneous AP firing rate were similar (0.4±0.1nM vs. 0.6±0.15nM, respectively). Moreover, the kinetics (t1/2) of the increases in PKA activity and spontaneous AP firing rate in response to ß-AR stimulation or PDE inhibition were tightly linked. We characterized the system rate-limiting biochemical reactions by integrating these experimentally derived data into a mechanistic-computational model. Model simulations predicted that phospholamban phosphorylation is a potent target of the increase in PKA activity that links to increase in spontaneous AP firing rate. In summary, the kinetics and stoichiometry of increases in PKA activity in response to a physiological (ß-AR stimulation) or pharmacological (PDE inhibitor) stimuli match those of changes in the AP firing rate. Thus Ca(2+)-cAMP/PKA-dependent phosphorylation limits the rate and magnitude of increase in spontaneous AP firing rate.


Subject(s)
Action Potentials/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Receptors, Adrenergic, beta/metabolism , Sinoatrial Node/drug effects , Animals , Calcium Signaling , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , Kinetics , Phosphodiesterase Inhibitors/administration & dosage , Phosphorylation/drug effects , Rabbits , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Signal Transduction/drug effects , Sinoatrial Node/metabolism , Sinoatrial Node/pathology
SELECTION OF CITATIONS
SEARCH DETAIL