Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
J Biol Chem ; 298(10): 102448, 2022 10.
Article in English | MEDLINE | ID: mdl-36063991

ABSTRACT

Mapping the self-organization and spatial distribution of membrane proteins is key to understanding their function. Developing methods that can provide insight into correlations between membrane protein colocalization and interactions is challenging. We report here on a correlated stochastic optical reconstruction microscopy/homoFRET imaging approach for resolving the nanoscale distribution and oligomeric state of membrane proteins. Using live cell homoFRET imaging of carcinoembryonic antigen-related cellular adhesion molecule 1, a cell-surface receptor known to exist in a complex equilibrium between monomer and dimer/oligomer states, we revealed highly heterogeneous diffraction-limited structures on the surface of HeLa cells. Furthermore, correlated super-resolved stochastic optical reconstruction microscopy imaging showed that these structures comprised a complex mixture and spatial distribution of self-associated carcinoembryonic antigen-related cellular adhesion molecule 1 molecules. In conclusion, this correlated approach provides a compelling strategy for addressing challenging questions about the interplay between membrane protein concentration, distribution, interaction, clustering, and function.


Subject(s)
Microscopy , Humans , HeLa Cells , Membrane Proteins , Microscopy/methods , Optical Imaging , Cytological Techniques
2.
Appl Opt ; 61(35): 10490-10498, 2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36607111

ABSTRACT

Digital holographic microscopy (DHM) has become an attractive imaging tool for the analysis of living cells and histological tissues. Telecentric DHM (TDHM) is a configuration of DHM that reduces the computational demands through a priori aberration corrections. However, TDHM requires a well-aligned optical pipeline to optimize its resolution and image quality (IQ), which has traditionally complicated the alignment process. Derived from optical interference functions, we offer here a set of methodologies to simplify TDHM design and alignment by determining the optimal +1-order position, which depends on the object-reference beam angle and the interference plane rotation angle. The methods are then experimentally tested and verified on a TDHM system by imaging living HeLa cells in suspension.


Subject(s)
Holography , Microscopy , Humans , Microscopy/methods , HeLa Cells , Holography/methods
3.
Development ; 144(7): 1350-1361, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28213553

ABSTRACT

Embryos extend their anterior-posterior (AP) axis in a conserved process known as axis elongation. Drosophila axis elongation occurs in an epithelial monolayer, the germband, and is driven by cell intercalation, cell shape changes, and oriented cell divisions at the posterior germband. Anterior germband cells also divide during axis elongation. We developed image analysis and pattern-recognition methods to track dividing cells from confocal microscopy movies in a generally applicable approach. Mesectoderm cells, forming the ventral midline, divided parallel to the AP axis, while lateral cells displayed a uniform distribution of division orientations. Mesectoderm cells did not intercalate and sustained increased AP strain before cell division. After division, mesectoderm cell density increased along the AP axis, thus relieving strain. We used laser ablation to isolate mesectoderm cells from the influence of other tissues. Uncoupling the mesectoderm from intercalating cells did not affect cell division orientation. Conversely, separating the mesectoderm from the anterior and posterior poles of the embryo resulted in uniformly oriented divisions. Our data suggest that mesectoderm cells align their division angle to reduce strain caused by mechanical forces along the AP axis of the embryo.


Subject(s)
Body Patterning , Cell Division , Cell Tracking/methods , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Animals , Automation , Biomechanical Phenomena , Cell Count , Cell Shape , Ectoderm/cytology , Mesoderm/cytology
4.
Proc Natl Acad Sci U S A ; 119(38): e2212419119, 2022 Sep 20.
Article in English | MEDLINE | ID: mdl-36070345
5.
J Biol Chem ; 293(38): 14723-14739, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30061153

ABSTRACT

Glycogen synthase kinase 3ß (GSK3ß) phosphorylates and thereby regulates a wide range of protein substrates involved in diverse cellular functions. Some GSK3ß substrates, such as c-Myc and Snail, are nuclear transcription factors, suggesting the possibility that GSK3ß function is controlled through its nuclear localization. Here, using ARPE-19 and MDA-MB-231 human cell lines, we found that inhibition of mTOR complex 1 (mTORC1) leads to partial redistribution of GSK3ß from the cytosol to the nucleus and to a GSK3ß-dependent reduction of the levels of both c-Myc and Snail. mTORC1 is known to be controlled by metabolic cues, such as by AMP-activated protein kinase (AMPK) or amino acid abundance, and we observed here that AMPK activation or amino acid deprivation promotes GSK3ß nuclear localization in an mTORC1-dependent manner. GSK3ß was detected on several distinct endomembrane compartments, including lysosomes. Consistently, disruption of late endosomes/lysosomes through a perturbation of RAS oncogene family member 7 (Rab7) resulted in loss of GSK3ß from lysosomes and in enhanced GSK3ß nuclear localization as well as GSK3ß-dependent reduction of c-Myc levels. These findings indicate that the nuclear localization and function of GSK3ß is suppressed by mTORC1 and suggest a link between metabolic conditions sensed by mTORC1 and GSK3ß-dependent regulation of transcriptional networks controlling cellular biomass production.


Subject(s)
Cell Nucleus/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mechanistic Target of Rapamycin Complex 1/physiology , Signal Transduction , AMP-Activated Protein Kinases/metabolism , Cell Compartmentation , Cell Line , Cell Line, Tumor , Cytosol/metabolism , Endosomes/metabolism , Glycogen Synthase Kinase 3 beta/chemistry , Humans , Intracellular Membranes/metabolism , Lysosomes/metabolism , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-myc/metabolism , Serine/metabolism
6.
J Cell Sci ; 130(15): 2579-2590, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28600323

ABSTRACT

Retromer is a multimeric protein complex that mediates endosome-to-trans-Golgi network (TGN) and endosome-to-plasma membrane trafficking of integral membrane proteins. Dysfunction of this complex has been linked to Alzheimer's disease and Parkinson's disease. The recruitment of retromer to endosomes is regulated by Rab7 (also known as RAB7A) to coordinate endosome-to-TGN trafficking of cargo receptor complexes. Rab7 is also required for the degradation of internalized integral membrane proteins, such as the epidermal growth factor receptor (EGFR). We found that Rab7 is palmitoylated and that this modification is not required for membrane anchoring. Palmitoylated Rab7 colocalizes efficiently with and has a higher propensity to interact with retromer than nonpalmitoylatable Rab7. Rescue of Rab7 knockout cells by expressing wild-type Rab7 restores efficient endosome-to-TGN trafficking, while rescue with nonpalmitoylatable Rab7 does not. Interestingly, Rab7 palmitoylation does not appear to be required for the degradation of EGFR or for its interaction with its effector, Rab-interacting lysosomal protein (RILP). Overall, our results indicate that Rab7 palmitoylation is required for the spatiotemporal recruitment of retromer and efficient endosome-to-TGN trafficking of the lysosomal sorting receptors.


Subject(s)
Endosomes/metabolism , Lipoylation , rab GTP-Binding Proteins/metabolism , trans-Golgi Network/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Endosomes/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Protein Transport , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins , trans-Golgi Network/genetics
7.
Mol Pharmacol ; 94(2): 917-925, 2018 08.
Article in English | MEDLINE | ID: mdl-29903751

ABSTRACT

Deletion of phenylalanine at position 508 (F508del) in cystic fibrosis transmembrane conductance regulator (CFTR) is the most common cystic fibrosis (CF)-causing mutation. Recently, ORKAMBI, a combination therapy that includes a corrector of the processing defect of F508del-CFTR (lumacaftor or VX-809) and a potentiator of channel activity (ivacaftor or VX-770), was approved for CF patients homozygous for this mutation. However, clinical studies revealed that the effect of ORKAMBI on lung function is modest and it was proposed that this modest effect relates to a negative impact of VX-770 on the stability of F508del-CFTR. In the current studies, we showed that this negative effect of VX-770 at 10 µM correlated with its inhibitory effect on VX-809-mediated correction of the interface between the second membrane spanning domain and the first nucleotide binding domain bearing F508del. Interestingly, we found that VX-770 exerted a similar negative effect on the stability of other membrane localized solute carriers (SLC26A3, SLC26A9, and SLC6A14), suggesting that this negative effect is not specific for F508del-CFTR. We determined that the relative destabilizing effect of a panel of VX-770 derivatives on F508del-CFTR correlated with their predicted lipophilicity. Polarized total internal reflection fluorescence microscopy on a supported lipid bilayer model shows that VX-770, and not its less lipophilic derivative, increased the fluidity of and reorganized the membrane. In summary, our findings show that there is a potential for nonspecific effects of VX-770 on the lipid bilayer and suggest that this effect may account for its destabilizing effect on VX-809- rescued F508del-CFTR.


Subject(s)
Aminophenols/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis/genetics , Quinolones/pharmacology , Sulfate Transporters/chemistry , Aminophenols/chemistry , Aminopyridines/pharmacology , Benzodioxoles/pharmacology , Cell Line , Cystic Fibrosis/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Freeze Drying , HEK293 Cells , Humans , Microscopy, Fluorescence , Mutation , Protein Stability/drug effects , Quinolones/chemistry
8.
J Biol Chem ; 292(34): 14292-14307, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28698382

ABSTRACT

Caveolae are bulb-shaped nanodomains of the plasma membrane that are enriched in cholesterol and sphingolipids. They have many physiological functions, including endocytic transport, mechanosensing, and regulation of membrane and lipid transport. Caveola formation relies on integral membrane proteins termed caveolins (Cavs) and the cavin family of peripheral proteins. Both protein families bind anionic phospholipids, but the precise roles of these lipids are unknown. Here, we studied the effects of phosphatidylserine (PtdSer), phosphatidylinositol 4-phosphate (PtdIns4P), and phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) on caveolar formation and dynamics. Using live-cell, single-particle tracking of GFP-labeled Cav1 and ultrastructural analyses, we compared the effect of PtdSer disruption or phosphoinositide depletion with caveola disassembly caused by cavin1 loss. We found that PtdSer plays a crucial role in both caveola formation and stability. Sequestration or depletion of PtdSer decreased the number of detectable Cav1-GFP puncta and the number of caveolae visualized by electron microscopy. Under PtdSer-limiting conditions, the co-localization of Cav1 and cavin1 was diminished, and cavin1 degradation was increased. Using rapamycin-recruitable phosphatases, we also found that the acute depletion of PtdIns4P and PtdIns(4,5)P2 has minimal impact on caveola assembly but results in decreased lateral confinement. Finally, we show in a model of phospholipid scrambling, a feature of apoptotic cells, that caveola stability is acutely affected by the scrambling. We conclude that the predominant plasmalemmal anionic lipid PtdSer is essential for proper Cav clustering, caveola formation, and caveola dynamics and that membrane scrambling can perturb caveolar stability.


Subject(s)
Caveolae/metabolism , Caveolin 1/metabolism , Cell Membrane/metabolism , Models, Biological , Phosphatidylserines/metabolism , RNA-Binding Proteins/metabolism , Animals , Caveolae/chemistry , Caveolae/ultrastructure , Caveolin 1/antagonists & inhibitors , Caveolin 1/chemistry , Caveolin 1/genetics , Cell Line , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cell Tracking , Cricetulus , Humans , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mesocricetus , Microscopy, Electron, Transmission , Microscopy, Video , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphatidylserines/chemistry , Protein Transport , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Time-Lapse Imaging
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(4): 447-457, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29343430

ABSTRACT

The mitochondrial glycerophospholipid cardiolipin plays important roles in mitochondrial biology. Most notably, cardiolipin directly binds to mitochondrial proteins and helps assemble and stabilize mitochondrial multi-protein complexes. Despite their importance for mitochondrial health, how the proteins involved in cardiolipin biosynthesis are organized and embedded in mitochondrial membranes has not been investigated in detail. Here we show that human PGS1 and CLS1 are constituents of large protein complexes. We show that PGS1 forms oligomers and associates with CLS1 and PTPMT1. Using super-resolution microscopy, we observed well-organized nanoscale structures formed by PGS1. Together with the observation that cardiolipin and CLS1 are not required for PGS1 to assemble in the complex we predict the presence of a PGS1-centered cardiolipin-synthesizing scaffold within the mitochondrial inner membrane. Using an unbiased proteomic approach we found that PGS1 and CLS1 interact with multiple cardiolipin-binding mitochondrial membrane proteins, including prohibitins, stomatin-like protein 2 and the MICOS components MIC60 and MIC19. We further mapped the protein-protein interaction sites between PGS1 and itself, CLS1, MIC60 and PHB. Overall, this study provides evidence for the presence of a cardiolipin synthesis structure that transiently interacts with cardiolipin-dependent protein complexes.


Subject(s)
Cardiolipins/biosynthesis , Cardiolipins/metabolism , Membrane Proteins/metabolism , Multienzyme Complexes/metabolism , Detergents/pharmacology , HEK293 Cells , Humans , Immunoprecipitation , Microscopy , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Prohibitins , Protein Binding/drug effects , Protein Multimerization/drug effects
10.
Biochim Biophys Acta Proteins Proteom ; 1865(11 Pt B): 1687-1695, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28844737

ABSTRACT

Identifying the key structural and dynamical determinants that drive the association of biomolecules, whether in solution, or perhaps more importantly in a membrane environment, has critical implications for our understanding of cellular dynamics, processes, and signaling. With recent advances in high-resolution imaging techniques, from the development of new molecular labels to technical advances in imaging methodologies and platforms, researchers are now reaping the benefits of being able to directly characterize and quantify local dynamics, structures, and conformations in live cells and tissues. These capabilities are providing unique insights into association stoichiometries, interactions, and structures on sub-micron length scales. We previously examined the role of lipid headgroup chemistry and phase state in guiding the formation of pseudoisocyanine (PIC) dye J-aggregates on supported planar bilayers [Langmuir, 25, 10719]. We describe here how these same J-aggregates can report on the in situ formation of organellar membrane domains in live cells. Live cell hyperspectral confocal microscopy using GFP-conjugated GTPase markers of early (Rab5) and late (Rab7) endosomes revealed that the PIC J-aggregates were confined to domains on either the limiting membrane or intralumenal vesicles (ILV) of late endosomes, known to be enriched in the anionic lipid bis(monoacylglycero)phosphate (BMP). Correlated confocal fluorescence - atomic force microscopy performed on endosomal membrane-mimetic supported planar lipid bilayers confirmed BMP-specific templating of the PIC J-aggregates. These data provide strong evidence for the formation of BMP-rich lipid domains during multivesicular body formation and portend the application of structured dye aggregates as markers of cellular membrane domain structure, size, and formation.


Subject(s)
Endosomes/metabolism , Glycerophosphates/metabolism , Intracellular Membranes/metabolism , Membrane Microdomains/metabolism , Monoglycerides/metabolism , Animals , CHO Cells , Cricetulus , Endosomes/ultrastructure , Hep G2 Cells , Humans , Intracellular Membranes/ultrastructure , Membrane Microdomains/ultrastructure , Mice , Microscopy, Atomic Force , Microscopy, Confocal , NIH 3T3 Cells , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
11.
J Biol Chem ; 289(15): 10419-10430, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24554723

ABSTRACT

The formation of fibrillar aggregates has long been associated with neurodegenerative disorders such as Alzheimer and Parkinson diseases. Although fibrils are still considered important to the pathology of these disorders, it is now widely understood that smaller amyloid oligomers are the toxic entities along the misfolding pathway. One characteristic shared by the majority of amyloid oligomers is the ability to disrupt membranes, a commonality proposed to be responsible for their toxicity, although the mechanisms linking this to cell death are poorly understood. Here, we describe the physical basis for the cytotoxicity of oligomers formed by the prion protein (PrP)-derived amyloid peptide PrP(106-126). We show that oligomers of this peptide kill several mammalian cells lines, as well as mouse cerebellar organotypic cultures, and we also show that they exhibit antimicrobial activity. Physical perturbation of model membranes mimicking bacterial or mammalian cells was investigated using atomic force microscopy, polarized total internal reflection fluorescence microscopy, and NMR spectroscopy. Disruption of anionic membranes proceeds through a carpet or detergent model as proposed for other antimicrobial peptides. By contrast, when added to zwitterionic membranes containing cholesterol-rich ordered domains, PrP(106-126) oligomers induce a loss of domain separation and decreased membrane disorder. Loss of raft-like domains may lead to activation of apoptotic pathways, resulting in cell death. This work sheds new light on the physical mechanisms of amyloid cytotoxicity and is the first to clearly show membrane type-specific modes of action for a cytotoxic peptide.


Subject(s)
Amyloid/chemistry , Peptide Fragments/chemistry , Prions/chemistry , Animals , Antimicrobial Cationic Peptides/chemistry , Cell Membrane/chemistry , Cerebellum/pathology , Cholesterol/chemistry , Detergents/chemistry , Escherichia coli/growth & development , Lipid Bilayers/chemistry , Lipids/chemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Organ Culture Techniques , PC12 Cells , Phosphatidylcholines/chemistry , Rats
12.
Biophys J ; 106(8): L29-31, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24739184

ABSTRACT

We use all-atom molecular dynamics simulations on a massive scale to compute the standard binding free energy of the 13-residue antimicrobial peptide indolicidin to a lipid bilayer. The analysis of statistical convergence reveals systematic sampling errors that correlate with reorganization of the bilayer on the microsecond timescale and persist throughout a total of 1.4 ms of sampling. Consistent with experimental observations, indolicidin induces membrane thinning, although the simulations significantly overestimate the lipophilicity of the peptide.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Lipid Bilayers/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemistry , Molecular Sequence Data , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Protein Binding
13.
Biochemistry ; 53(37): 5885-94, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25171271

ABSTRACT

TAR DNA binding protein of 43 kDa (TDP-43) has been implicated in the pathogenesis of a broad range of neurodegenerative diseases termed TDP-43 proteinopathies, which encompass a spectrum of diseases ranging from amyotrophic lateral sclerosis to frontotemporal dementia. Pathologically misfolded and aggregated forms of TDP-43 are found in cytoplasmic inclusion bodies of affected neurons in these diseases. The mechanism by which TDP-43 misfolding causes disease is not well-understood. Current hypotheses postulate that the TDP-43 aggregation process plays a major role in pathogenesis. We amplify that hypothesis and suggest that binding of cognate ligands to TDP-43 can stabilize the native functional state of the protein and ameliorate aggregation. We expressed recombinant TDP-43 containing an N-terminal Venus yellow fluorescent protein tag in Escherichia coli and induced its aggregation by altering solvent salt concentrations and examined the extent to which various oligonucleotide molecules affect its aggregation in vitro using aggregation-induced turbidity assays. We show that vYFP-TDP-43 binding to its naturally occurring RNA target that comprises a sequence on the 3'UTR region of its mRNA improves its solubility, suggesting interplay among TDP-43 solubility, oligonucleotide binding, and TDP-43 autoregulation.


Subject(s)
3' Untranslated Regions , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , RNA, Messenger/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Protein Multimerization , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
J Biol Chem ; 288(41): 29654-69, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24005674

ABSTRACT

Cell-cell contacts are fundamental to multicellular organisms and are subject to exquisite levels of control. The carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) can engage in both cis-homophilic (parallel) oligomerization and trans-homophilic (anti-parallel) binding. In this study, we establish that the CEACAM1 transmembrane domain has a propensity to form cis-dimers via the transmembrane-embedded (432)GXXXG(436) motif and that this basal state is overcome when activated calmodulin binds to the CEACAM1 cytoplasmic domain. Although mutation of the (432)GXXXG(436) motif reduced CEACAM1 oligomerization, it did not affect surface localization of the receptor or influence CEACAM1-dependent cellular invasion by the pathogenic Neisseria. The mutation did, however, have a striking effect on CEACAM1-dependent cellular aggregation, increasing both the kinetics of cell-cell association and the size of cellular aggregates formed. CEACAM1 association with tyrosine kinase c-Src and tyrosine phosphatases SHP-1 and SHP-2 was not affected by the (432)GXXXG(436) mutation, consistent with their association with the monomeric form of wild type CEACAM1. Collectively, our results establish that a dynamic oligomer-to-monomer shift in surface-expressed CEACAM1 facilitates trans-homophilic binding and downstream effector signaling.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Protein Multimerization , Signal Transduction , Amino Acid Motifs/genetics , Amino Acid Sequence , Antigens, CD/genetics , Calcium/metabolism , Calmodulin/metabolism , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Membrane/metabolism , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , src-Family Kinases/metabolism
15.
Biochim Biophys Acta ; 1828(10): 2272-82, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23500349

ABSTRACT

Direct correlation of molecular conformation with local structure is critical to studies of protein- and peptide-membrane interactions, particularly in the context of membrane-facilitated aggregation, and disruption or disordering. Infrared spectroscopy has long been a mainstay for determining molecular conformation, following folding dynamics, and characterizing reactions. While tremendous advances have been made in improving the spectral and temporal resolution of infrared spectroscopy, it has only been with the introduction of scanned-probe techniques that exploit the raster-scanning tip as either a source, scattering tool, or measurement probe that researchers have been able to obtain sub-diffraction limit IR spectra. This review will examine the history of correlated scanned-probe IR spectroscopies, from their inception to their use in studies of molecular aggregates, membrane domains, and cellular structures. The challenges and opportunities that these platforms present for examining dynamic phenomena will be discussed. This article is part of a Special Issue entitled: FTIR in membrane proteins and peptide studies.


Subject(s)
Biophysics , Cell Membrane , Spectroscopy, Fourier Transform Infrared/methods , Microscopy/methods
16.
Biochim Biophys Acta ; 1828(11): 2757-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23954588

ABSTRACT

Sticholysins (Sts) I and II (StI/II) are pore-forming toxins (PFTs) produced by the Caribbean Sea anemone Stichodactyla helianthus belonging to the actinoporin family, a unique class of eukaryotic PFTs exclusively found in sea anemones. The role of lipid phase co-existence in the mechanism of the action of membranolytic proteins and peptides is not clearly understood. As for actinoporins, it has been proposed that phase separation promotes pore forming activity. However little is known about the effect of sticholysins on the phase separation of lipids in membranes. To gain insight into the mechanism of action of sticholysins, we evaluated the effect of these proteins on lipid segregation using differential scanning calorimetry (DSC) and atomic force microscopy (AFM). New evidence was obtained reflecting that these proteins reduce line tension in the membrane by promoting lipid mixing. In terms of the relevance for the mechanism of action of actinoporins, we hypothesize that expanding lipid disordered phases into lipid ordered phases decreases the lipid packing at the borders of the lipid raft, turning it into a more suitable environment for N-terminal insertion and pore formation.


Subject(s)
Cnidarian Venoms/pharmacology , Lipids/chemistry , Membrane Microdomains/metabolism , Sea Anemones/metabolism , Animals , Calorimetry, Differential Scanning , Microscopy, Atomic Force , Organic Chemicals/pharmacology , Sea Anemones/chemistry
17.
J Am Chem Soc ; 136(45): 15849-52, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25350268

ABSTRACT

Here we describe for the first time the synthesis of colloidally stable, brightly luminescent perfluorodecyl-capped silicon nanocrystals and compare the properties of solutions and films made from them with those of their perhydrodecyl-capped relatives. The perfluorodecyl capping group compared to the perhydrodecyl capping group yields superior hydrophobicity and much greater resistance to air oxidation, the enhanced electron-withdrawing character induces blue shifts in the wavelength of photoluminescence, and the lower-frequency carbon-fluorine stretching modes disfavor non-radiative relaxation pathways and boost the absolute photoluminescence quantum yield. Together these attributes bode well for advanced materials and biomedical applications founded upon perfluorodecyl-protected silicon nanocrystals.


Subject(s)
Fluorocarbons/chemistry , Luminescent Agents/chemistry , Nanoparticles/chemistry , Silicon/chemistry , Luminescent Measurements , Oxidation-Reduction , Wettability
18.
HardwareX ; 17: e00508, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38327674

ABSTRACT

We present the design of a low-cost, portable telecentric digital holographic microscope (P-TDHM) that utilizes off-the-shelf components. We describe the system's hardware and software elements and evaluate its performance by imaging samples ranging from nano-printed targets to live HeLa cells, HEK293 cells, and Dolichospermum via both in-line and off-axis modes. Our results demonstrate that the system can acquire high quality quantitative phase images with nanometer axial and sub-micron lateral resolution in a small form factor, making it a promising candidate for resource-limited settings and remote locations. Our design represents a significant step forward in making telecentric digital holographic microscopy accessible and affordable to the broader community.

19.
Nat Commun ; 15(1): 3120, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600106

ABSTRACT

Salmonella utilizes a type 3 secretion system to translocate virulence proteins (effectors) into host cells during infection1. The effectors modulate host cell machinery to drive uptake of the bacteria into vacuoles, where they can establish an intracellular replicative niche. A remarkable feature of Salmonella invasion is the formation of actin-rich protuberances (ruffles) on the host cell surface that contribute to bacterial uptake. However, the membrane source for ruffle formation and how these bacteria regulate membrane mobilization within host cells remains unclear. Here, we show that Salmonella exploits membrane reservoirs for the generation of invasion ruffles. The reservoirs are pre-existing tubular compartments associated with the plasma membrane (PM) and are formed through the activity of RAB10 GTPase. Under normal growth conditions, membrane reservoirs contribute to PM homeostasis and are preloaded with the exocyst subunit EXOC2. During Salmonella invasion, the bacterial effectors SipC, SopE2, and SopB recruit exocyst subunits from membrane reservoirs and other cellular compartments, thereby allowing exocyst complex assembly and membrane delivery required for bacterial uptake. Our findings reveal an important role for RAB10 in the establishment of membrane reservoirs and the mechanisms by which Salmonella can exploit these compartments during host cell invasion.


Subject(s)
Salmonella Infections , Salmonella typhimurium , Humans , Salmonella typhimurium/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Salmonella Infections/microbiology , Cell Membrane/metabolism , Membranes/metabolism , HeLa Cells
20.
Cell Rep ; 43(7): 114417, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38980795

ABSTRACT

The ability to sense and respond to osmotic fluctuations is critical for the maintenance of cellular integrity. We used gene co-essentiality analysis to identify an unappreciated relationship between TSC22D2, WNK1, and NRBP1 in regulating cell volume homeostasis. All of these genes have paralogs and are functionally buffered for osmo-sensing and cell volume control. Within seconds of hyperosmotic stress, TSC22D, WNK, and NRBP family members physically associate into biomolecular condensates, a process that is dependent on intrinsically disordered regions (IDRs). A close examination of these protein families across metazoans revealed that TSC22D genes evolved alongside a domain in NRBPs that specifically binds to TSC22D proteins, which we have termed NbrT (NRBP binding region with TSC22D), and this co-evolution is accompanied by rapid IDR length expansion in WNK-family kinases. Our study reveals that TSC22D, WNK, and NRBP genes evolved in metazoans to co-regulate rapid cell volume changes in response to osmolarity.

SELECTION OF CITATIONS
SEARCH DETAIL