Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
NPJ Parkinsons Dis ; 9(1): 137, 2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37741841

ABSTRACT

Alpha synuclein (a-syn) is an intrinsically disordered protein prevalent in neurons, and aggregated forms are associated with synucleinopathies including Parkinson's disease (PD). Despite the biomedical importance and extensive studies, the physiological role of a-syn and its participation in etiology of PD remain uncertain. We showed previously in model RBL cells that a-syn colocalizes with mitochondrial membranes, depending on formation of N-terminal helices and increasing with mitochondrial stress1. We have now characterized this colocalization and functional correlates in RBL, HEK293, and N2a cells. We find that expression of a-syn enhances stimulated mitochondrial uptake of Ca2+ from the ER, depending on formation of its N-terminal helices but not on its disordered C-terminal tail. Our results are consistent with a-syn acting as a tether between mitochondria and ER, and we show increased contacts between these two organelles using structured illumination microscopy. We tested mitochondrial stress caused by toxins related to PD, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP/MPP+) and carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and found that a-syn prevents recovery of stimulated mitochondrial Ca2+ uptake. The C-terminal tail, and not N-terminal helices, is involved in this inhibitory activity, which is abrogated when phosphorylation site serine-129 is mutated (S129A). Correspondingly, we find that MPTP/MPP+ and CCCP stress is accompanied by both phosphorylation (pS129) and aggregation of a-syn. Overall, our results indicate that a-syn can participate as a tethering protein to modulate Ca2+ flux between ER and mitochondria, with potential physiological significance. A-syn can also prevent cellular recovery from toxin-induced mitochondrial dysfunction, which may represent a pathological role of a-syn in the etiology of PD.

2.
bioRxiv ; 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37163091

ABSTRACT

Alpha synuclein (a-syn) is an intrinsically disordered protein prevalent in neurons, and aggregated forms are associated with synucleinopathies including Parkinson' disease (PD). Despite the biomedical importance and extensive studies, the physiological role of a-syn and its participation in etiology of PD remain uncertain. We showed previously in model RBL cells that a-syn colocalizes with mitochondrial membranes, depending on formation of N-terminal helices and increasing with mitochondrial stress. 1 We have now characterized this colocalization and functional correlates in RBL, HEK293, and N2a cells. We find that expression of a-syn enhances stimulated mitochondrial uptake of Ca 2+ from the ER, depending on formation of its N-terminal helices but not on its disordered C-terminal tail. Our results are consistent with a-syn acting as a tether between mitochondria and ER, and we show increased contacts between these two organelles using structured illumination microscopy. We tested mitochondrial stress caused by toxins related to PD, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP/MPP+) and carbonyl cyanide m-chlorophenyl hydrazone (CCCP), and found that a-syn prevents recovery of stimulated mitochondrial Ca 2+ uptake. The C-terminal tail, and not N-terminal helices, is involved in this inhibitory activity, which is abrogated when phosphorylation site serine-129 is mutated (S129A). Correspondingly, we find that MPTP/MPP+ and CCCP stress is accompanied by both phosphorylation (pS129) and aggregation of a-syn. Overall, our results indicate that a-syn can participate as a tethering protein to modulate Ca 2+ flux between ER and mitochondria, with potential physiological significance. A-syn can also prevent cellular recovery from toxin-induced mitochondrial dysfunction, which may represent a pathological role of a-syn in the etiology of PD.

3.
Biomolecules ; 12(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36551244

ABSTRACT

Alpha-synuclein is a presynaptic protein linked to Parkinson's disease with a poorly characterized physiological role in regulating the synaptic vesicle cycle. Using RBL-2H3 cells as a model system, we earlier reported that wild-type alpha-synuclein can act as both an inhibitor and a potentiator of stimulated exocytosis in a concentration-dependent manner. The inhibitory function is constitutive and depends on membrane binding by the helix-2 region of the lipid-binding domain, while potentiation becomes apparent only at high concentrations. Using structural and functional characterization of conformationally selective mutants via a combination of spectroscopic and cellular assays, we show here that binding affinity for isolated vesicles similar in size to synaptic vesicles is a primary determinant of alpha-synuclein-mediated potentiation of vesicle release. Inhibition of release is sensitive to changes in the region linking the helix-1 and helix-2 regions of the N-terminal lipid-binding domain and may require some degree of coupling between these regions. Potentiation of release likely occurs as a result of alpha-synuclein interactions with undocked vesicles isolated away from the active zone in internal pools. Consistent with this, we observe that alpha-synuclein can disperse vesicles from in vitro clusters organized by condensates of the presynaptic protein synapsin-1.


Subject(s)
Parkinson Disease , Synaptic Membranes , Synaptic Vesicles , alpha-Synuclein , Humans , alpha-Synuclein/chemistry , Lipids/chemistry , Parkinson Disease/metabolism , Synaptic Vesicles/metabolism , Protein Domains , Synaptic Membranes/chemistry
4.
J Phys Chem B ; 126(12): 2325-2336, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35294838

ABSTRACT

Plasma membranes host numerous receptors, sensors, and ion channels involved in cellular signaling. Phase separation within the plasma membrane has emerged as a key biophysical regulator of signaling reactions in multiple physiological and pathological contexts. There is much evidence that plasma membrane composition supports the coexistence of liquid-ordered (Lo) and liquid-disordered (Ld) phases or domains at physiological conditions. However, this phase/domain separation is nanoscopic and transient in live cells. It has been recently proposed that transbilayer coupling between the inner and outer leaflets of the plasma membrane is driven by their asymmetric lipid distribution and by dynamic cytoskeleton-lipid composites that contribute to the formation and transience of Lo/Ld phase separation in live cells. In this Perspective, we highlight new approaches to investigate how transbilayer coupling may influence phase separation. For quantitative evaluation of the impact of these interactions, we introduce an experimental strategy centered around Imaging Fluorescence Correlation Spectroscopy (ImFCS), which measures membrane diffusion with very high precision. To demonstrate this strategy, we choose two well-established model systems for transbilayer interactions: cross-linking by multivalent antigen of immunoglobulin E bound to receptor FcεRI and cross-linking by cholera toxin B of GM1 gangliosides. We discuss emerging methods to systematically perturb membrane lipid composition, particularly exchange of outer leaflet lipids with exogenous lipids using methyl alpha cyclodextrin. These selective perturbations may be quantitatively evaluated with ImFCS and other high-resolution biophysical tools to discover novel principles of lipid-mediated phase separation in live cells in the context of their pathophysiological relevance.


Subject(s)
Membrane Lipids , Cell Membrane/chemistry , Diffusion , Membrane Lipids/metabolism , Spectrometry, Fluorescence
5.
Methods Mol Biol ; 2421: 1-19, 2022.
Article in English | MEDLINE | ID: mdl-34870808

ABSTRACT

Cell surface receptors that bind the Fc segment of antibodies to initiate signaling play fundamental roles in immune responses. Multiple, diverse Fc receptors (e.g., Fc gamma, Fc-alpha, and Fc-epsilon) are expressed on different immune cells, including natural killer cells, macrophages, mast cells, and neutrophils. Fc receptors bind particular antibody isotypes (e.g., IgG, IgA, IgE, respectively) thereby sensitizing the cells to their specific antigens. Receptor clustering by antigen or other multivalent ligands induces a signaling cascade that leads to targeted secretion of chemical mediators (e.g., histamine, cytokines, and chemokines) and other cell-specific responses. Spatial targeting and compartmentalization are common mechanisms for regulating Fc receptor signaling. However, the tools for studying these dynamic interactions at cellular levels have been limited due to the nanoscale dimensions of the signaling complexes and their dispersal across the cell surface. To overcome these limitations in our model system, we use microfabricated surfaces containing spatially defined ligands to cluster and activate IgE receptors (FcεRI), which initiate allergic responses by mast cells. Micron-scale control of receptor assemblies allows investigation with conventional fluorescence microscopy of spatially regulated redistributions of intracellular signaling components. This approach in conjunction with biochemical techniques has proven valuable for investigating immune receptor signaling.


Subject(s)
Receptors, Fc/immunology , Antigens , Ligands , Mast Cells , Phagocytosis , Receptors, IgE
6.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34433665

ABSTRACT

Antigen (Ag) crosslinking of immunoglobulin E-receptor (IgE-FcεRI) complexes in mast cells stimulates transmembrane (TM) signaling, requiring phosphorylation of the clustered FcεRI by lipid-anchored Lyn tyrosine kinase. Previous studies showed that this stimulated coupling between Lyn and FcεRI occurs in liquid ordered (Lo)-like nanodomains of the plasma membrane and that Lyn binds directly to cytosolic segments of FcεRI that it initially phosphorylates for amplified activity. Net phosphorylation above a nonfunctional threshold is achieved in the stimulated state but not in the resting state, and current evidence supports the hypothesis that this relies on Ag crosslinking to disrupt a balance between Lyn and tyrosine phosphatase activities. However, the structural interactions that underlie the stimulation process remain poorly defined. This study evaluates the relative contributions and functional importance of different types of interactions leading to suprathreshold phosphorylation of Ag-crosslinked IgE-FcεRI in live rat basophilic leukemia mast cells. Our high-precision diffusion measurements by imaging fluorescence correlation spectroscopy on multiple structural variants of Lyn and other lipid-anchored probes confirm subtle, stimulated stabilization of the Lo-like nanodomains in the membrane inner leaflet and concomitant sharpening of segregation from liquid disordered (Ld)-like regions. With other structural variants, we determine that lipid-based interactions are essential for access by Lyn, leading to phosphorylation of and protein-based binding to clustered FcεRI. By contrast, TM tyrosine phosphatase, PTPα, is excluded from these regions due to its Ld-preference and steric exclusion of TM segments. Overall, we establish a synergy of lipid-based, protein-based, and steric interactions underlying functional TM signaling in mast cells.


Subject(s)
Antigens/metabolism , Cell Membrane/metabolism , Lipids/physiology , Mast Cells/metabolism , Receptors, IgE/metabolism , Signal Transduction , Animals , Antigens/immunology , CHO Cells , Cell Line, Tumor , Cells, Cultured , Cricetulus , Green Fluorescent Proteins/metabolism , Lipid Metabolism , Mast Cells/immunology , Nanostructures , Rats , src-Family Kinases/metabolism
7.
Protein J ; 39(5): 476-486, 2020 10.
Article in English | MEDLINE | ID: mdl-33211253

ABSTRACT

Epidermal growth factor receptor (EGFR) dysregulation is observed in many human cancers and is both a cause of oncogenesis and a target for chemotherapy. We previously showed that partial charge neutralization of the juxtamembrane (JX) region of EGFR via the EGFR R1-6 mutant construct induces constitutive receptor activation and transformation of NIH 3T3 cells, both from the plasma membrane and from the ER when combined with the ER-retaining L417H mutation (Bryant et al. in J Biol Chem 288:34930-34942, 2013). Here, we use chemical crosslinking and immunoblotting to show that these mutant constructs form constitutive, phosphorylated dimers in both the plasma membrane and the ER. Furthermore, we combine this electrostatic perturbation with conformationally-restricted receptor mutants to provide evidence that activation of EGFR R1-6 dimers requires functional coupling both between the EGFR extracellular dimerization arms and between intracellular tyrosine kinase domains. These findings provide evidence that the electrostatic charge of the JX region normally serves as a negative regulator of functional dimerization of EGFR.


Subject(s)
Protein Multimerization , Amino Acid Substitution , Animals , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mice , Mutation, Missense , NIH 3T3 Cells , Phosphorylation , Protein Domains
9.
Mol Biol Cell ; 31(7): 709-723, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31895009

ABSTRACT

A myriad of transient, nanoscopic lipid- and protein-based interactions confer a steady-state organization of the plasma membrane in resting cells that is poised to orchestrate assembly of key signaling components upon reception of an extracellular stimulus. Although difficult to observe directly in live cells, these subtle interactions can be discerned by their impact on the diffusion of membrane constituents. Here, we quantified the diffusion properties of a panel of structurally distinct lipid, lipid-anchored, and transmembrane (TM) probes in RBL mast cells by imaging fluorescence correlation spectroscopy (ImFCS). We developed a statistical analysis of data combined from many pixels over multiple cells to characterize differences in diffusion coefficients as small as 10%, which reflect differences in underlying interactions. We found that the distinctive diffusion properties of lipid probes can be explained by their dynamic partitioning into Lo-like proteolipid nanodomains, which encompass a major fraction of the membrane and whose physical properties are influenced by actin polymerization. Effects on diffusion of functional protein modules in both lipid--anchored and TM probes reflect additional complexity in steady state membrane organization. The contrast we observe between different probes diffusing through the same membrane milieu represents the dynamic resting steady state, which serves as a baseline for monitoring plasma membrane remodeling that occurs upon stimulation.


Subject(s)
Cell Membrane/metabolism , Mast Cells/metabolism , Spectrometry, Fluorescence , Actins/metabolism , Animals , Cell Line , Diffusion , Green Fluorescent Proteins/metabolism , Imaging, Three-Dimensional , Immunoglobulin E/metabolism , Lipids/chemistry , Polymerization , Rats , Receptors, IgE/metabolism
10.
NPJ Parkinsons Dis ; 5: 12, 2019.
Article in English | MEDLINE | ID: mdl-31263746

ABSTRACT

We characterized phenotypes in RBL-2H3 mast cells transfected with human alpha synuclein (a-syn) using stimulated exocytosis of recycling endosomes as a proxy for similar activities of synaptic vesicles in neurons. We found that low expression of a-syn inhibits stimulated exocytosis and that higher expression causes slight enhancement. NMR measurements of membrane interactions correlate with these functional effects: they are eliminated differentially by mutants that perturb helical structure in the helix 1 (A30P) or NAC/helix-2 (V70P) regions of membrane-bound a-syn, but not by other PD-associated mutants or C-terminal truncation. We further found that a-syn (but not A30P or V70P mutants) associates weakly with mitochondria, but this association increases markedly under conditions of cellular stress. These results highlight the importance of specific structural features of a-syn in regulating vesicle release, and point to a potential role for a-syn in perturbing mitochondrial function under pathological conditions.

11.
Biol Open ; 7(9)2018 Sep 27.
Article in English | MEDLINE | ID: mdl-30097519

ABSTRACT

Lipid phase heterogeneity in plasma membranes is thought to play a key role in targeting cellular signaling, but efforts to test lipid raft and related hypotheses are limited by the spatially dynamic nature of these phase-based structures in cells and by experimental characterization tools. We suggest that perturbation of plasma membrane structure by lipid derivatives offers a general method for assessing functional roles for ordered lipid regions in membrane and cell biology. We previously reported that short chain ceramides with either C2 or C6 acyl chains inhibit antigen-stimulated Ca2+ mobilization (Gidwani et al., 2003). We now show that these short chain ceramides inhibit liquid order (Lo)-liquid disorder (Ld) phase separation in giant plasma membrane vesicles that normally occurs at low temperatures. Furthermore, they are effective inhibitors of tyrosine phosphorylation stimulated by antigen, as well as store-operated Ca2+ entry. In Jurkat T cells, C6-ceramide is also effective at inhibiting Ca2+ mobilization stimulated by either anti-TCR or thapsigargin, consistent with the view that these short chain ceramides effectively interfere with functional responses that depend on ordered lipid regions in the plasma membrane.

12.
Stem Cells ; 36(10): 1603-1616, 2018 10.
Article in English | MEDLINE | ID: mdl-29938858

ABSTRACT

The role of lipid metabolism in epithelial stem cell (SC) function and carcinogenesis is poorly understood. The transcription factor Runx1 is known to regulate proliferation in mouse epithelial hair follicle (HF) SCs in vivo and in several mouse and human epithelial cancers. We found a novel subset of in vivo Runx1 HFSC target genes related to lipid metabolism and demonstrated changes in distinct classes of lipids driven by Runx1. Inhibition of lipid-enzymes Scd1 and Soat1 activity synergistically reduces proliferation of mouse skin epithelial cells and of human skin and oral squamous cell carcinoma cultured lines. Varying Runx1 levels induces changes in skin monounsaturated fatty acids (e.g., oleate, a product of Scd1) as shown by our lipidome analysis. Furthermore, varying Runx1 levels, the inhibition of Scd1, or the addition of Scd1-product oleate, individually affects the plasma membrane organization (or fluidity) in mouse keratinocytes. These factors also affect the strength of signal transduction through the membranes for Wnt, a pathway that promotes epithelial (cancer) cell proliferation and HFSC activation. Our working model is that HFSC factor Runx1 modulates the fatty acid production, which affects membrane organization, facilitating signal transduction for rapid proliferation of normal and cancer epithelial cells. Stem Cells 2018;36:1603-1616.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Epithelial Cells/metabolism , Stearoyl-CoA Desaturase/metabolism , Sterol O-Acyltransferase/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Core Binding Factor Alpha 2 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/genetics , Epithelial Cells/cytology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Lipid Metabolism/genetics , Mice , Mice, Knockout , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Stearoyl-CoA Desaturase/genetics , Stem Cells/cytology , Stem Cells/metabolism , Sterol O-Acyltransferase/genetics , Transfection
13.
J Phys Chem B ; 122(13): 3500-3513, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29432021

ABSTRACT

Lipid phase heterogeneity in the plasma membrane is thought to be crucial for many aspects of cell signaling, but the physical basis of participating membrane domains such as "lipid rafts" remains controversial. Here we consider a lattice model yielding a phase diagram that includes several states proposed to be relevant for the cell membrane, including microemulsion-which can be related to membrane curvature-and Ising critical behavior. Using a neural-network-based machine learning approach, we compute the full phase diagram of this lattice model. We analyze selected regions of this phase diagram in the context of a signaling initiation event in mast cells: recruitment of the membrane-anchored tyrosine kinase Lyn to a cluster of transmembrane IgE-FcεRI receptors. We find that model membrane systems in microemulsion and Ising critical states can mediate roughly equal levels of kinase recruitment (binding energy ∼ -0.6 kB T), whereas a membrane near a tricritical point can mediate a much stronger kinase recruitment (-1.7 kB T). By comparing several models for lipid heterogeneity within a single theoretical framework, this work points to testable differences between existing models. We also suggest the tricritical point as a new possibility for the basis of membrane domains that facilitate preferential partitioning of signaling components.


Subject(s)
Lipids/chemistry , Molecular Dynamics Simulation , Proteins/chemistry , Monte Carlo Method
14.
Sci Rep ; 7(1): 15586, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29138425

ABSTRACT

The high-affinity receptor for IgE expressed on the surface of mast cells and basophils interacts with antigens, via bound IgE antibody, and triggers secretion of inflammatory mediators that contribute to allergic reactions. To understand how past inputs (memory) influence future inflammatory responses in mast cells, a microfluidic device was used to precisely control exposure of cells to alternating stimulatory and non-stimulatory inputs. We determined that the response to subsequent stimulation depends on the interval of signaling quiescence. For shorter intervals of signaling quiescence, the second response is blunted relative to the first response, whereas longer intervals of quiescence induce an enhanced second response. Through an iterative process of computational modeling and experimental tests, we found that these memory-like phenomena arise from a confluence of rapid, short-lived positive signals driven by the protein tyrosine kinase Syk; slow, long-lived negative signals driven by the lipid phosphatase Ship1; and slower degradation of Ship1 co-factors. This work advances our understanding of mast cell signaling and represents a generalizable approach for investigating the dynamics of signaling systems.


Subject(s)
Inflammation/immunology , Mast Cells/immunology , Receptors, IgE/immunology , Signal Transduction/immunology , Animals , Antibodies/immunology , Antigens/immunology , Basophils/immunology , Humans , Inflammation/genetics , Inflammation/metabolism , Lab-On-A-Chip Devices , Mast Cells/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/immunology , Receptors, IgE/genetics , Signal Transduction/genetics , Syk Kinase/genetics , Syk Kinase/immunology
15.
Mol Biol Cell ; 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28794269

ABSTRACT

We examined the spatial targeting of early and downstream signaling mediated by the IgE receptor (FcεRI) in RBL mast cells utilizing surface-patterned 2,4 dinitrophenyl (DNP) ligands. Micron-sized features of DNP are presented as densely immobilized conjugates of bovine serum albumin (DNP-BSA) or mobile in a supported lipid bilayer (DNP-SLB). Although soluble anti-DNP IgE binds uniformly across features for both pattern types, IgE bound to FcεRI on cells shows distinctive distributions: uniform for DNP-SLB and edge-concentrated for DNP-BSA. These distributions of IgE-FcεRI propagate to the spatial recruitment of early signaling proteins, including spleen tyrosine kinase (Syk), linker for activation of T cells (LAT), and activated phospholipase C gamma 1 (PLCγ1), which all localize with engaged receptors. We found stimulated polymerization of F-actin is not required for Syk recruitment but is progressively involved in the recruitment of LAT and PLCγ1. We further found ß1- and ß3-integrins colocalize with IgE-FcεRI at patterned ligand surfaces as cells spread. This recruitment corresponds to directed exocytosis of recycling endosomes (REs) containing these integrins and their fibronectin ligand. Together, our results show targeting of signaling components, including integrins, to regions of clustered IgE-FcεRI in processes that depend on stimulated actin polymerization and outward trafficking of REs.

16.
J Cell Sci ; 130(17): 2821-2832, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28724757

ABSTRACT

Decreased luminal endoplasmic reticulum (ER) Ca2+ concentration triggers oligomerization and clustering of the ER Ca2+ sensor STIM1 to promote its association with plasma membrane Orai1 Ca2+ channels leading to increased Ca2+ influx. A key step in STIM1 activation is the release of its SOAR domain from an intramolecular clamp formed with the STIM1 first coiled-coil (CC1) region. Using a truncated STIM1(1-343) molecule that captures or releases the isolated SOAR domain depending on luminal ER Ca2+ concentrations, we analyzed the early molecular events that control the intramolecular clamp formed between the CC1 and SOAR domains. We found that STIM1 forms constitutive dimers, and its CC1 domain can bind the SOAR domain of another STIM1 molecule in trans. Artificial oligomerization failed to liberate the SOAR domain or activate STIM1 unless the luminal Ca2+-sensing domains were removed. We propose that the release of SOAR from its CC1 interaction is controlled by changes in the orientation of the two CC1 domains in STIM1 dimers. Ca2+ unbinding in the STIM1 luminal domains initiates the conformational change allowing SOAR domain liberation and clustering, leading to Orai1 channel activation.


Subject(s)
Protein Multimerization , Stromal Interaction Molecule 1/chemistry , Stromal Interaction Molecule 1/metabolism , Animals , COS Cells , Cell Survival , Chlorocebus aethiops , Imaging, Three-Dimensional , Mutation/genetics , Protein Conformation , Protein Domains , Protein Stability , Stromal Interaction Molecule 1/genetics
19.
Biochim Biophys Acta Biomembr ; 1859(9 Pt A): 1430-1435, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28024796

ABSTRACT

The cell surface receptor for epidermal growth factor (EGFR), a receptor tyrosine kinase, is a key player in normal cell growth and proliferation. Mutations in this receptor often lead to oncological transformation and other pathologies. Because of its representation of the receptor tyrosine kinase family and its important role in health and disease, a broad range of studies have been carried out in many laboratories to investigate the structural basis for transmembrane receptor activation and the resulting assembly of cytosolic signaling components. This review highlights two approaches our laboratory has taken to gain more detailed information about both aspects: Surface patterned ligands to examine recruitment of the signaling machinery, and mutational analysis to examine the regulatory role of EGFR's juxtamembrane segment. This article is part of a Special Issue entitled: Interactions between membrane receptors in cellular membranes edited by Kalina Hristova.


Subject(s)
Cell Membrane/genetics , Cell Proliferation/genetics , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , Cell Membrane/metabolism , DNA Mutational Analysis , Humans , Ligands , Mutation , Protein Binding , Signal Transduction
20.
Chem ; 1(2): 273-286, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27891534

ABSTRACT

Graphene oxide (GO) has attracted intense interest for use in living systems and environmental applications. GO's compatibility with mammalian cells is sometimes inferred from its low cytotoxicity, but such conclusions ignore non-lethal effects that will influence GO's utility. Here we demonstrate, with rat basophilic leukemia (RBL) cells, profound plasma membrane (PM) ruffling and shedding induced by GO using confocal and live cell fluorescence microscopy, as well as scanning electron microscopy. These membrane structures contain immunoglobulin E receptors, are resistant to detergents, and lack detectable fluorescence labeling of F-actin and fibronectin. The formation of these membrane structures correlates with a loss of contact inhibition between RBL cells. We observe similar cellular responses towards GO for NIH-3T3 fibroblast cells and MDA-MB-231 human breast cancer cells. These findings reveal a previously unreported cellular response towards foreign nanomaterials. Membrane ruffling and shedding raise fundamental questions about how GO interacts with the PM, as well as its potential to modulate cellular mechanosensing for tissue engineering, stem cell differentiation, and other biomedical applications.

SELECTION OF CITATIONS
SEARCH DETAIL
...