Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 7831, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36539423

ABSTRACT

Protein micropatterning enables robust control of cell positioning on electron-microscopy substrates for cryogenic electron tomography (cryo-ET). However, the combination of regulated cell boundaries and the underlying electron-microscopy substrate (EM-grids) provides a poorly understood microenvironment for cell biology. Because substrate stiffness and morphology affect cellular behavior, we devised protocols to characterize the nanometer-scale details of the protein micropatterns on EM-grids by combining cryo-ET, atomic force microscopy, and scanning electron microscopy. Measuring force displacement characteristics of holey carbon EM-grids, we found that their effective spring constant is similar to physiological values expected from skin tissues. Despite their apparent smoothness at light-microscopy resolution, spatial boundaries of the protein micropatterns are irregular at nanometer scale. Our protein micropatterning workflow provides the means to steer both positioning and morphology of cell doublets to determine nanometer details of punctate adherens junctions. Our workflow serves as the foundation for studying the fundamental structural changes governing cell-cell signaling.


Subject(s)
Image Processing, Computer-Assisted , Proteins , Image Processing, Computer-Assisted/methods , Cryoelectron Microscopy/methods , Carbon/chemistry , Signal Transduction
2.
Mol Biol Cell ; 33(14): br28, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36287913

ABSTRACT

Matrix stiffness and dimensionality have been shown to be major determinants of cell behavior. However, a workflow for examining nanometer-scale responses of the associated molecular machinery is not available. Here, we describe a comprehensive, quantitative workflow that permits the analysis of cells responding to mechanical and dimensionality cues in their native state at nanometer scale by cryogenic electron tomography. Using this approach, we quantified distinct cytoskeletal nanoarchitectures and vesicle phenotypes induced in human mammary epithelial cells in response to stiffness and dimensionality of reconstituted basement membrane. Our workflow closely recapitulates the microenvironment associated with acinar morphogenesis and identified distinct differences in situ at nanometer scale. Using drug treatment, we showed that molecular events and nanometer-scale rearrangements triggered by engagement of apical cell receptors with reconstituted basement membrane correspond to changes induced by reduction of cortical tension. Our approach is fully adaptable to any kind of stiffness regime, extracellular matrix composition, and drug treatment.


Subject(s)
Epithelial Cells , Extracellular Matrix , Humans , Workflow , Morphogenesis , Extracellular Matrix/metabolism , Electron Microscope Tomography
3.
EMBO J ; 41(17): e109205, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35880301

ABSTRACT

Patient-derived organoids and cellular spheroids recapitulate tissue physiology with remarkable fidelity. We investigated how engagement with a reconstituted basement membrane in three dimensions (3D) supports the polarized, stress resilient tissue phenotype of mammary epithelial spheroids. Cells interacting with reconstituted basement membrane in 3D had reduced levels of total and actin-associated filamin and decreased cortical actin tension that increased plasma membrane protrusions to promote negative plasma membrane curvature and plasma membrane protein associations linked to protein secretion. By contrast, cells engaging a reconstituted basement membrane in 2D had high cortical actin tension that forced filamin unfolding and endoplasmic reticulum (ER) associations. Enhanced filamin-ER interactions increased levels of PKR-like ER kinase effectors and ER-plasma membrane contact sites that compromised calcium homeostasis and diminished cell viability. Consequently, cells with decreased cortical actin tension had reduced ER stress and survived better. Consistently, cortical actin tension in cellular spheroids regulated polarized basement membrane membrane deposition and sensitivity to exogenous stress. The findings implicate cortical actin tension-mediated filamin unfolding in ER function and underscore the importance of tissue mechanics in organoid homeostasis.


Subject(s)
Actins , Endoplasmic Reticulum , Actins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Epithelial Cells/metabolism , Filamins/metabolism , Phenotype
4.
J Struct Biol ; 213(4): 107801, 2021 12.
Article in English | MEDLINE | ID: mdl-34582983

ABSTRACT

With the rapid increase and accessibility of high-resolution imaging technologies of cells, the interpretation of results relies more and more on the assumption that the three-dimensional integrity of the surrounding cellular landscape is not compromised by the experimental setup. However, the only available technology for directly probing the structural integrity of whole-cell preparations at the nanoscale is electron cryo-tomography, which is time-consuming, costly, and complex. We devised an accessible, inexpensive and reliable screening assay to quickly report on the compatibility of experimental protocols with preserving the structural integrity of whole-cell preparations at the nanoscale. Our Rapid Cell Integrity Assessment (RCIA) assay is executed at room temperature and relies solely on light microscopy imaging. Using cellular electron cryo-tomography as a benchmark, we verify that RCIA accurately unveils the adverse impact of reagents and/or protocols such as those used for virus inactivation or to arrest dynamic processes on the cellular nanoarchitecture.


Subject(s)
Cryoelectron Microscopy/methods , Electron Microscope Tomography/methods , Eukaryotic Cells/ultrastructure , Imaging, Three-Dimensional/methods , Nanostructures/ultrastructure , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/ultrastructure , Animals , Cells, Cultured , Eukaryotic Cells/chemistry , Eukaryotic Cells/classification , HeLa Cells , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/ultrastructure , Mice , Microscopy, Fluorescence/methods , Mitochondria/chemistry , Mitochondria/ultrastructure , NIH 3T3 Cells , Nanostructures/chemistry , Reproducibility of Results , THP-1 Cells
5.
J Micromech Microeng ; 29(11)2019 Nov.
Article in English | MEDLINE | ID: mdl-32879557

ABSTRACT

Cryogenic electron tomography is the highest resolution tool available for structural analysis of macromolecular organization inside cells. Micropatterning of extracellular matrix (ECM) proteins is an established in vitro cell culture technique used to control cell shape. Recent traction force microscopy studies have shown correlation between cell morphology and the regulation of force transmission. However, it remains unknown how cells sustain increased strain energy states and localized stresses at the supramolecular level. Here, we report a technology to enable direct observation of mesoscale organization in epithelial cells under morphological modulation, using a maskless protein photopatterning method (PRIMO) to confine cells to ECM micropatterns on electron microscopy substrates. These micropatterned cell culture substrates can be used in mechanobiology research to correlate changes in nanometer-scale organization at cell-cell and cell-ECM contacts to strain energy states and traction stress distribution in the cell.

6.
J Cell Biol ; 193(2): 347-63, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21502359

ABSTRACT

Although RII protein kinase A (PKA) regulatory subunits are constitutively localized to discrete cellular compartments through binding to A-kinase-anchoring proteins (AKAPs), RI subunits are primarily diffuse in the cytoplasm. In this paper, we report a novel AKAP-dependent localization of RIα to distinct organelles, specifically, multivesicular bodies (MVBs). This localization depends on binding to AKAP11, which binds tightly to free RIα or RIα in complex with catalytic subunit (holoenzyme). However, recruitment to MVBs occurs only with the release of PKA catalytic subunit (PKAc). This recruitment is reversed by reassociation with PKAc, and it is disrupted by the presence of AKAP peptides, mutations in the RIα AKAP-binding site, or knockdown of AKAP11. Cyclic adenosine monophosphate binding not only unleashes active PKAc but also leads to the targeting of AKAP11:RIα to MVBs. Therefore, we show that the RIα holoenzyme is part of a signaling complex with AKAP11, in which AKAP11 may direct RIα functionality after disassociation from PKAc. This model defines a new paradigm for PKA signaling.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Multivesicular Bodies , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Amino Acid Sequence , Binding Sites/genetics , Catalytic Domain , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Isoenzymes/metabolism , Molecular Sequence Data , Mutation , Protein Binding , Protein Transport , Signal Transduction
7.
Cold Spring Harb Protoc ; 2011(1): pdb.prot5547, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21205847

ABSTRACT

Correlation of real-time or time-lapse light microscopy (LM) with electron microscopy (EM) of cells can be performed with biarsenical dyes. These dyes fluorescently label tetracysteine-tagged proteins so that they can be imaged with LM and, upon fluorescent photoconversion of 3,3'-diaminobenzidine tetrahydrochloride (DAB), with EM as well. In the following protocol, cells expressing tetracysteine-tagged proteins are labeled for 1 h with biarsenical dyes. The volumes indicated are for a single 30-mm culture dish containing 2 mL of labeling medium. Scale the suggested volumes up or down depending upon the size of the culture dish used in the labeling. The same procedure can be adapted for longer labeling times by lowering the amount of dye used to 50-100 nM; however, the amount of the competing dithiol EDT is maintained at 10-20 µM. Longer labeling times often produce higher signal-to-noise ratios and cause less trauma to the treated cells prior to imaging.


Subject(s)
Recombinant Proteins/analysis , Staining and Labeling/methods , Arsenicals , Cell Line , Coloring Agents , Image Processing, Computer-Assisted/methods , Microscopy/methods
8.
Cold Spring Harb Protoc ; 2011(1): pdb.prot5548, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21205848

ABSTRACT

Correlated light microscopy (LM)/electron microscopy (EM) analysis can be achieved by using biarsenical dyes to fluorescently label tetracysteine-tagged proteins. Once live cell imaging using LM is complete, cellular activity can be halted promptly using a glutaraldehyde-based fixative. Rapid fixation preserves cellular ultrastructure and limits diffusion of reaction products. This protocol provides details on rapid fixation of cells, followed by fluorescence photoconversion of 3,3'-diaminobenzidine tetrahydrochloride (DAB) and sample processing for EM that can be correlated with the live cell LM images.


Subject(s)
Recombinant Proteins/analysis , Staining and Labeling/methods , Arsenicals , Cell Line , Fluorescent Dyes , Image Processing, Computer-Assisted/methods , Microscopy, Electron/methods
9.
Cold Spring Harb Protoc ; 2011(1): pdb.top94, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21205860

ABSTRACT

Fundamental to obtaining a more complete understanding of the roles played by macromolecular complexes in cells is the ability to map their location, movement, and transient interactions at high temporal and high spatial resolution. Unfortunately, probes capable of allowing direct correlation of real-time or time-lapse light microscopy (LM) with electron microscopic observations are relatively few. Genetically encoded fluorescent reporters such as green fluorescent protein (GFP) have revolutionized live cell imaging studies but are not directly visible by electron microscopy (EM). Fluorescent nanoparticles or quantum dots are a type of label for LM that can also be visualized directly by EM, but targeting these to cytoplasmic proteins in living cells remains difficult. One method that does allow for highly correlated LM and EM with excellent preservation of cellular ultrastructure is fluorescence photoconversion, in which a fluorescent compound causes the deposition of a reaction product that can be rendered electron-dense and directly visualized by EM. We have used this method in combination with a class of genetically encoded peptide tags that can be labeled in living cells by fluorophores bearing two appropriately spaced arsenic atoms (biarsenicals). The tetracysteine motif is short, easily inserted into or attached to the termini of the host protein, and can be used in combination with other molecular tags such as GFP and its derivatives. This article presents methods to label cells with biarsenicals, conduct live cell imaging recording sessions, and generate specimens that can be evaluated by EM for a correlated LM/EM analysis.


Subject(s)
Arsenicals/metabolism , Cysteine/metabolism , Microscopy/methods , Staining and Labeling/methods , Arsenicals/chemistry , Cysteine/chemistry
10.
Nat Protoc ; 5(10): 1666-77, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20885379

ABSTRACT

In this paper, we provide a general protocol for labeling proteins with the membrane-permeant fluorogenic biarsenical dye fluorescein arsenical hairpin binder-ethanedithiol (FlAsH-EDT2). Generation of the tetracysteine-tagged protein construct by itself is not described, as this is a protein-specific process. This method allows site-selective labeling of proteins in living cells and has been applied to a wide variety of proteins and biological problems. We provide here a generally applicable labeling procedure and discuss the problems that can occur as well as general considerations that must be taken into account when designing and implementing the procedure. The method can even be applied to proteins with expression below 1 pmol mg⁻¹ of protein, such as G protein-coupled receptors, and it can be used to study the intracellular localization of proteins as well as functional interactions in fluorescence resonance energy transfer experiments. The labeling procedure using FlAsH-EDT2 as described takes 2-3 h, depending on the number of samples to be processed.


Subject(s)
Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Organometallic Compounds/chemistry , Proteins/chemistry , Staining and Labeling/methods , Amino Acid Sequence , Animals , Arsenicals/chemistry , Binding Sites , Cells, Cultured , Fluorescence Resonance Energy Transfer , Humans , Protein Binding , Proteins/analysis
11.
Mol Microbiol ; 76(1): 173-89, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20149103

ABSTRACT

The bacterium Caulobacter crescentus has morphologically and functionally distinct cell poles that undergo sequential changes during the cell cycle. We show that the PopZ oligomeric network forms polar ribosome exclusion zones that change function during cell cycle progression. The parS/ParB chromosomal centromere is tethered to PopZ at one pole prior to the initiation of DNA replication. During polar maturation, the PopZ-centromere tether is broken, and the PopZ zone at that pole then switches function to act as a recruitment factor for the ordered addition of multiple proteins that promote the transformation of the flagellated pole into a stalked pole. Stalked pole assembly, in turn, triggers the initiation of chromosome replication, which signals the formation of a new PopZ zone at the opposite cell pole, where it functions to anchor the newly duplicated centromere that has traversed the long axis of the cell. We propose that pole-specific control of PopZ function co-ordinates polar development and cell cycle progression by enabling independent assembly and tethering activities at the two cell poles.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/physiology , Cell Cycle , Cell Polarity , Caulobacter crescentus/metabolism , Centromere/metabolism , Chromosomes, Bacterial/metabolism , DNA Replication , DNA, Bacterial/metabolism , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microscopy, Immunoelectron , Models, Biological , Models, Molecular , Protein Multimerization
12.
Exp Eye Res ; 88(3): 600-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19073179

ABSTRACT

Mutant connexins have been linked to hereditary congenital cataracts. One such mutant causes a proline-to-serine substitution at position 88 in human connexin 50 (CX50P88S). In transfected cells, CX50P88S does not form gap junctions, but localizes in cytoplasmic multilamellar structures. We studied the dynamics of formation and the stability of these structures in HeLa cells stably transfected with CX50P88S containing a tetracysteine motif appended to its C-terminus (HeLa-CX50P88S(Cys)(4) cells). The tetracysteine motif binds the membrane-permeable biarsenical compounds, FlAsH and ReAsH, which become fluorescent upon binding allowing detection of CX50P88S(Cys)(4) by fluorescence microscopy or by transmission electron microscopy after the ReAsH-driven fluorescent photoconversion of diaminobenzidine. CX50P88S structures were long-lived. Pulse labeling of HeLa-CX50P88S(Cys)(4) cells with FlAsH followed by a chase and ReAsH labeling showed a differential distribution of the labels, with older CX50P88S surrounded by newly synthesized protein. Formation of CX50P88S accumulations was not affected by treatments that block ER-to-Golgi transport. Transmission electron microscopy and tomographic reconstruction revealed that CX50P88S accumulations corresponded to closely apposed circular or semicircular membrane stacks that were sometimes continuous with the rough endoplasmic reticulum. These results suggest that CX50P88S accumulations originate from the rough endoplasmic reticulum and that mutant protein is sequentially added resulting in long-lived cytoplasmic particles. The persistence of these particles in the lens may cause light scattering and the pulverulent cataracts observed in affected individuals.


Subject(s)
Cataract/genetics , Connexins/genetics , Cytoplasm/metabolism , Eye Proteins/genetics , Mutation , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Connexins/metabolism , Cytoplasm/ultrastructure , Electron Microscope Tomography , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Eye Proteins/metabolism , HeLa Cells , Humans , Microscopy, Electron
13.
J Struct Biol ; 161(3): 439-46, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17998167

ABSTRACT

Virus assembly occurs in a complex environment and is dependent upon viral and cellular components being properly correlated in time and space. The simplicity of the flock house virus (FHV) capsid and the extensive structural, biochemical and genetic characterization of the virus make it an excellent system for studying in vivo virus assembly. The tetracysteine motif (CCPGCC), that induces fluorescence in bound biarsenical compounds (FlAsH and ReAsH), was genetically inserted in the coat protein, to visualize this gene product during virus infection. The small size of this modification when compared to those made by traditional fluorescent proteins minimizes disruption of the coat proteins numerous functions. ReAsH not only fluoresces when bound to the tetracysteine motif but also allows correlated electron microscopy (EM) of the same cell following photoconversion and osmium staining. These studies demonstrated that the coat protein was concentrated in discrete patches in the cell. High pressure freezing (HPF) followed by freeze substitution (FS) of infected cells showed that these patches were formed by virus particles in crystalline arrays. EM tomography (EMT) of the HPF/FS prepared samples showed that these arrays were proximal to highly modified mitochondria previously established to be the site of RNA replication. Two features of the mitochondrial modification are approximately 60 nm spherules that line the outer membrane and the large chamber created by the convolution induced in the entire organelle.


Subject(s)
Capsid/ultrastructure , Drosophila/virology , Nodaviridae/ultrastructure , Virus Assembly/physiology , Animals , Luminescent Proteins , Microscopy, Electron , Mitochondria/ultrastructure , Tomography
14.
J Biol Chem ; 282(43): 31733-43, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17715132

ABSTRACT

Pannexins are newly discovered channel proteins expressed in many different tissues and abundantly in the vertebrate central nervous system. Based on membrane topology, folding and secondary structure prediction, pannexins are proposed to form gap junction-like structures. We show here that Pannexin1 forms a hexameric channel and reaches the cell surface but, unlike connexins, is N-glycosylated. Using site-directed mutagenesis we analyzed three putative N-linked glycosylation sites and examined the effects of each mutation on channel expression. We show for the first time that Pannexin1 is glycosylated at Asn-254 and that this residue is important for plasma membrane targeting. The glycosylation of Pannexin1 at its extracellular surface makes it unlikely that two oligomers could dock to form an intercellular channel. Ultrastructural analysis by electron microscopy confirmed that Pannexin1 junctional areas do not appear as canonical gap junctions. Rather, Pannexin1 channels are distributed throughout the plasma membrane. We propose that N-glycosylation of Pannexin1 could be a significant mechanism for regulating the trafficking of these membrane proteins to the cell surface in different tissues.


Subject(s)
Cell Membrane/metabolism , Connexins/chemistry , Connexins/metabolism , Ion Channels/ultrastructure , Asparagine/metabolism , Biotinylation , Cell Line , Cell Membrane/ultrastructure , Connexins/genetics , Connexins/ultrastructure , Cross-Linking Reagents/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Glycosylation , Humans , Immunohistochemistry , Kidney/cytology , Models, Biological , Mutation , Nerve Tissue Proteins , Protein Processing, Post-Translational , Protein Structure, Secondary , Succinimides/pharmacology
15.
Biochem J ; 408(3): 375-85, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17714073

ABSTRACT

The C-terminus of the most abundant and best-studied gap-junction protein, connexin43, contains multiple phosphorylation sites and protein-binding domains that are involved in regulation of connexin trafficking and channel gating. It is well-documented that SDS/PAGE of NRK (normal rat kidney) cell lysates reveals at least three connexin43-specific bands (P0, P1 and P2). P1 and P2 are phosphorylated on multiple, unidentified serine residues and are found primarily in gap-junction plaques. In the present study we prepared monoclonal antibodies against a peptide representing the last 23 residues at the C-terminus of connexin43. Immunofluorescence studies showed that one antibody (designated CT1) bound primarily to connexin43 present in the Golgi apparatus, whereas the other antibody (designated IF1) labelled predominately connexin43 present in gap junctions. CT1 immunoprecipitates predominantly the P0 form whereas IF1 recognized all three bands. Peptide mapping, mutational analysis and protein-protein interaction experiments revealed that unphosphorylated Ser364 and/or Ser365 are critical for CT1 binding. The IF1 paratope binds to residues Pro375-Asp379 and requires Pro375 and Pro377. These proline residues are also necessary for ZO-1 interaction. These studies indicate that the conformation of Ser364/Ser365 is important for intracellular localization, whereas the tertiary structure of Pro375-Asp379 is essential in targeting and regulation of gap junctional connexin43.


Subject(s)
Antibodies/immunology , Connexin 43/chemistry , Gap Junctions/chemistry , Golgi Apparatus/chemistry , Animals , Cell Line , Connexin 43/genetics , Connexin 43/immunology , Dogs , Electrophoresis, Polyacrylamide Gel , Immunoprecipitation , Microscopy, Confocal , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Phosphorylation , Protein Conformation , Rats
17.
Proc Natl Acad Sci U S A ; 103(47): 17777-82, 2006 Nov 21.
Article in English | MEDLINE | ID: mdl-17101980

ABSTRACT

Combinations of molecular tags visible in light and electron microscopes become particularly advantageous in the analysis of dynamic cellular components like the Golgi apparatus. This organelle disassembles at the onset of mitosis and, after a sequence of poorly understood events, reassembles after cytokinesis. The precise location of Golgi membranes and resident proteins during mitosis remains unclear, partly due to limitations of molecular markers and the resolution of light microscopy. We generated a fusion consisting of the first 117 residues of alpha-mannosidase II tagged with a fluorescent protein and a tetracysteine motif. The mannosidase component guarantees docking into the Golgi membrane, with the tags exposed in the lumen. The fluorescent protein is optically visible without further treatment, whereas the tetracysteine tag can be reduced acutely with a membrane-permeant phosphine, labeled with ReAsH, monitored in the light microscope, and used to trigger the photoconversion of diaminobenzidine, allowing 4D optical recording on live cells and correlated ultrastructural analysis by electron microscopy. These methods reveal that Golgi reassembly is preceded by the formation of four colinear clusters at telophase, two per daughter cell. Within each daughter, the smaller cluster near the midbody gradually migrates to rejoin the major cluster on the far side of the nucleus and asymmetrically reconstitutes a single Golgi apparatus, first in one daughter cell and then in the other. Our studies provide previously undescribed insights into Golgi disassociation and reassembly during mitosis and offer a powerful approach to follow recombinant protein distribution in 4D imaging and correlated high-resolution analysis.


Subject(s)
Golgi Apparatus , Microscopy, Electron/methods , Mitosis/physiology , Staining and Labeling/methods , Cysteine/genetics , Cysteine/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , HeLa Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Microscopy, Electron/instrumentation , Oxidation-Reduction , Peptides/genetics , Peptides/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , alpha-Mannosidase/genetics , alpha-Mannosidase/metabolism
18.
J Biol Chem ; 281(12): 7994-8009, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16407179

ABSTRACT

Single site mutations in connexins have provided insights about the influence specific amino acids have on gap junction synthesis, assembly, trafficking, and functionality. We have discovered a single point mutation that eliminates functionality without interfering with gap junction formation. The mutation occurs at a threonine residue located near the cytoplasmic end of the third transmembrane helix. This threonine is strictly conserved among members of the alpha- and beta-connexin subgroups but not the gamma-subgroup. In HeLa cells, connexin43 and connexin26 mutants are synthesized, traffic to the plasma membrane, and make gap junctions with the same overall appearance as wild type. We have isolated connexin26T135A gap junctions both from HeLa cells and baculovirus-infected insect Sf9 cells. By using cryoelectron microscopy and correlation averaging, difference images revealed a small but significant size change within the pore region and a slight rearrangement of the subunits between mutant and wild-type connexons expressed in Sf9 cells. Purified, detergent-solubilized mutant connexons contain both hexameric and partially disassembled structures, although wild-type connexons are almost all hexameric, suggesting that the three-dimensional mutant connexon is unstable. Mammalian cells expressing gap junction plaques composed of either connexin43T154A or connexin26T135A showed an absence of dye coupling. When expressed in Xenopus oocytes, these mutants, as well as a cysteine substitution mutant of connexin50 (connexin50T157C), failed to produce electrical coupling in homotypic and heteromeric pairings with wild type in a dominant-negative effect. This mutant may be useful as a tool for knocking down or knocking out connexin function in vitro or in vivo.


Subject(s)
Cell Membrane/metabolism , Connexins/chemistry , Connexins/genetics , Mutation , Threonine/chemistry , Amino Acid Sequence , Animals , Baculoviridae/metabolism , Cell Line , Connexin 26 , Connexin 43/genetics , Cryoelectron Microscopy , Cysteine/chemistry , Cytoplasm/metabolism , DNA, Complementary/metabolism , Electrophysiology , Fluorescent Dyes/pharmacology , Gap Junctions , Genes, Dominant , HeLa Cells , Humans , Image Processing, Computer-Assisted , Insecta , Keratinocytes/metabolism , Light , Microscopy, Electron , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/metabolism , Oxygen/metabolism , Phylogeny , Point Mutation , RNA, Complementary/metabolism , Rats , Time Factors , Transfection , Xenopus
19.
J Biol Chem ; 281(14): 9667-76, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16434405

ABSTRACT

A mutation linked to autistic spectrum disorders encodes an Arg to Cys replacement in the C-terminal portion of the extracellular domain of neuroligin-3. The solvent-exposed Cys causes virtually complete retention of the protein in the endoplasmic reticulum when the protein is expressed in transfected cells. An identical Cys substitution was reported for butyrylcholinesterase through genotyping patients with post-succinylcholine apnea. Neuroligin, butyrylcholinesterase, and acetylcholinesterase are members of the alpha,beta-hydrolase fold family of proteins sharing sequence similarity and common tertiary structures. Although these proteins have distinct oligomeric assemblies and cellular dispositions, homologous Arg residues in neuroligin-3 (Arg-451), in butyrylcholinesterase (Arg-386), and in acetylcholinesterase (Arg-395) are conserved in all studied mammalian species. To examine whether an homologous Arg to Cys mutation affects related proteins similarly despite their differing capacities to oligomerize, we inserted homologous mutations in the acetylcholinesterase and butyrylcholinesterase cDNAs. Using confocal fluorescence microscopy and analysis of oligosaccharide processing, we find that the homologous Arg to Cys mutation also results in endoplasmic reticulum retention of the two cholinesterases. Small quantities of mutated acetylcholinesterase exported from the cell retain activity but show a greater K(m), a much smaller k(cat), and altered substrate inhibition. The nascent proteins associate with chaperones during processing, but the mutation presumably restricts processing through the endoplasmic reticulum and Golgi apparatus, because of local protein misfolding and inability to oligomerize. The mutation may alter the capacity of these proteins to dissociate from their chaperone prior to oligomerization and processing for export.


Subject(s)
Acetylcholinesterase/genetics , Autistic Disorder/genetics , Butyrylcholinesterase/genetics , Endoplasmic Reticulum/physiology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Folding , Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cell Adhesion Molecules, Neuronal , DNA, Complementary , Membrane Proteins/metabolism , Microscopy, Confocal , Molecular Chaperones , Mutagenesis, Site-Directed , Nerve Tissue Proteins/metabolism , Point Mutation , Protein Structure, Tertiary
20.
Nat Methods ; 2(3): 171-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15782185

ABSTRACT

Fluorescence resonance energy transfer (FRET) from cyan to yellow fluorescent proteins (CFP/YFP) is a well-established method to monitor protein-protein interactions or conformational changes of individual proteins. But protein functions can be perturbed by fusion of large tags such as CFP and YFP. Here we use G protein-coupled receptor (GPCR) activation in living cells as a model system to compare YFP with the small, membrane-permeant fluorescein derivative with two arsen-(III) substituents (fluorescein arsenical hairpin binder; FlAsH) targeted to a short tetracysteine sequence. Insertion of CFP and YFP into human adenosine A(2A) receptors allowed us to use FRET to monitor receptor activation but eliminated coupling to adenylyl cyclase. The CFP/FlAsH-tetracysteine system gave fivefold greater agonist-induced FRET signals, similar kinetics (time constant of 66-88 ms) and perfectly normal downstream signaling. Similar results were obtained for the mouse alpha(2A)-adrenergic receptor. Thus, FRET from CFP to FlAsH reports GPCR activation in living cells without disturbing receptor function and shows that the small size of the tetracysteine-biarsenical tag can be decisively advantageous.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , GTP-Binding Proteins/metabolism , Kidney/metabolism , Protein Interaction Mapping/methods , Receptors, G-Protein-Coupled/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Fluorescent Dyes , Green Fluorescent Proteins , HeLa Cells , Humans , Receptor, Adenosine A2A
SELECTION OF CITATIONS
SEARCH DETAIL
...