Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Traffic ; 21(6): 419-429, 2020 06.
Article in English | MEDLINE | ID: mdl-32246734

ABSTRACT

Once secretory proteins have been targeted to the endoplasmic reticulum (ER) lumen, the proteins typically remain partitioned from the cytosol. If the secretory proteins misfold, they can be unfolded and retrotranslocated into the cytosol for destruction by the proteasome by ER-Associated protein Degradation (ERAD). Here, we report that correctly folded and targeted luminal ER fluorescent protein reporters accumulate in the cytosol during acute misfolded secretory protein stress in yeast. Photoactivation fluorescence microscopy experiments reveal that luminal reporters already localized to the ER relocalize to the cytosol, even in the absence of essential ERAD machinery. We named this process "ER reflux." Reflux appears to be regulated in a size-dependent manner for reporters. Interestingly, prior heat shock stress also prevents ER stress-induced reflux. Together, our findings establish a new ER stress-regulated pathway for relocalization of small luminal secretory proteins into the cytosol, distinct from the ERAD and preemptive quality control pathways. Importantly, our results highlight the value of fully characterizing the cell biology of reporters and describe a simple modification to maintain luminal ER reporters in the ER during acute ER stress.


Subject(s)
Cytosol/metabolism , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Protein Folding , Endoplasmic Reticulum-Associated Degradation
2.
Proc Natl Acad Sci U S A ; 117(1): 522-531, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871169

ABSTRACT

Interleukin 15 (IL-15) is an essential cytokine for the survival and proliferation of natural killer (NK) cells. IL-15 activates signaling by the ß and common γ (γc) chain heterodimer of the IL-2 receptor through trans-presentation by cells expressing IL-15 bound to the α chain of the IL-15 receptor (IL-15Rα). We show here that membrane-associated IL-15Rα-IL-15 complexes are transferred from presenting cells to NK cells through trans-endocytosis and contribute to the phosphorylation of ribosomal protein S6 and NK cell proliferation. NK cell interaction with soluble or surface-bound IL-15Rα-IL-15 complex resulted in Stat5 phosphorylation and NK cell survival at a concentration or density of the complex much lower than required to stimulate S6 phosphorylation. Despite this efficient response, Stat5 phosphorylation was reduced after inhibition of metalloprotease-induced IL-15Rα-IL-15 shedding from trans-presenting cells, whereas S6 phosphorylation was unaffected. Conversely, inhibition of trans-endocytosis by silencing of the small GTPase TC21 or expression of a dominant-negative TC21 reduced S6 phosphorylation but not Stat5 phosphorylation. Thus, trans-endocytosis of membrane-associated IL-15Rα-IL-15 provides a mode of regulating NK cells that is not afforded to IL-2 and is distinct from activation by soluble IL-15. These results may explain the strict IL-15 dependence of NK cells and illustrate how the cellular compartment in which receptor-ligand interaction occurs can influence functional outcome.


Subject(s)
Cell Proliferation , Dendritic Cells/metabolism , Interleukin-15 Receptor alpha Subunit/metabolism , Interleukin-15/metabolism , Killer Cells, Natural/physiology , Cell Communication/physiology , Cell Line , Endocytosis/physiology , Healthy Volunteers , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Phosphorylation/physiology , Primary Cell Culture , Ribosomal Protein S6/metabolism
3.
Mol Biol Cell ; 30(13): 1575-1577, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31194669

ABSTRACT

Each faculty recruiting season, many postdocs ask, "What is a chalk talk?" The chalk talk is many things-a sales pitch, a teaching demonstration, a barrage of questions, and a description of a future research program. The chalk talk is arguably the most important component of a faculty search interview. Yet few postdocs or grad students receive training or practice in giving a chalk talk. In the following essay, I'll cover the basics of chalk talk design and preparation.


Subject(s)
Teaching/education , Communication , Humans , Research Design , Students , Teaching/trends
4.
Nat Commun ; 9(1): 5368, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30560927

ABSTRACT

Foxp3+CD4+ regulatory T (Treg) cells are essential for preventing fatal autoimmunity and safeguard immune homeostasis in vivo. While expression of the transcription factor Foxp3 and IL-2 signals are both required for the development and function of Treg cells, the commitment to the Treg cell lineage occurs during thymic selection upon T cell receptor (TCR) triggering, and precedes the expression of Foxp3. Whether signals beside TCR contribute to establish Treg cell epigenetic and functional identity is still unknown. Here, using a mouse model with reduced IL-2 signaling, we show that IL-2 regulates the positioning of the pioneer factor SATB1 in CD4+ thymocytes and controls genome wide chromatin accessibility of thymic-derived Treg cells. We also show that Treg cells receiving only low IL-2 signals can suppress endogenous but not WT autoreactive T cell responses in vitro and in vivo. Our findings have broad implications for potential therapeutic strategies to reprogram Treg cells in vivo.


Subject(s)
Cellular Reprogramming/genetics , Epigenesis, Genetic/immunology , Interleukin-2/metabolism , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/genetics , Cell Differentiation/immunology , Cellular Reprogramming/immunology , Disease Models, Animal , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Listeriosis/immunology , Listeriosis/microbiology , Male , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Thymocytes/immunology , Thymocytes/metabolism , Thymocytes/physiology , Thymus Gland/cytology
5.
Sci Rep ; 8(1): 14738, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30283009

ABSTRACT

The ability of fluorescent proteins (FPs) to fold robustly is fundamental to the autocatalytic formation of the chromophore. While the importance of the tertiary protein structure is well appreciated, the impact of individual amino acid mutations for FPs is often not intuitive and requires direct testing. In this study, we describe the engineering of a monomeric photoswitchable FP, moxMaple3, for use in oxidizing cellular environments, especially the eukaryotic secretory pathway. Surprisingly, a point mutation to replace a cysteine substantially improved the yield of correctly folded FP capable of chromophore formation, regardless of cellular environment. The improved folding of moxMaple3 increases the fraction of visibly tagged fusion proteins, as well as FP performance in PALM super-resolution microscopy, and thus makes moxMaple3 a robust monomeric FP choice for PALM and optical highlighting applications.


Subject(s)
Cysteine/chemistry , Eukaryotic Cells/metabolism , Green Fluorescent Proteins/chemistry , Luminescent Proteins/chemistry , Amino Acids/chemistry , Green Fluorescent Proteins/genetics , Humans , Luminescent Proteins/genetics , Microscopy, Fluorescence/methods , Oxidation-Reduction , Protein Folding , Protein Structure, Tertiary/genetics
6.
Biophys J ; 115(7): 1146-1155, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30219286

ABSTRACT

The saga of fluorescence recovery after photobleaching (FRAP) illustrates how disparate technical developments impact science. Starting with the classic 1976 Axelrod et al. work in Biophysical Journal, FRAP (originally fluorescence photobleaching recovery) opened the door to extraction of quantitative information from photobleaching experiments, laying the experimental and theoretical groundwork for quantifying both the mobility and the mobile fraction of a labeled population of proteins. Over the ensuing years, FRAP's reach dramatically expanded, with new developments in GFP technology and turn-key confocal microscopy, which enabled measurement of protein diffusion and binding/dissociation rates in virtually every compartment within the cell. The FRAP technique and data catalyzed an exchange of ideas between biophysicists studying membrane dynamics, cell biologists focused on intracellular dynamics, and systems biologists modeling the dynamics of cell activity. The outcome transformed the field of cellular biology, leading to a fundamental rethinking of long-held theories of cellular dynamism. Here, we review the pivotal FRAP studies that made these developments and conceptual changes possible, which gave rise to current models of complex cell dynamics.


Subject(s)
Fluorescence Recovery After Photobleaching/methods , Movement , Proteins/metabolism , Cell Membrane/metabolism , Organelles/metabolism , Protein Transport
7.
Elife ; 62017 07 28.
Article in English | MEDLINE | ID: mdl-28753126

ABSTRACT

Like all other secretory proteins, the HIV-1 envelope glycoprotein gp160 is targeted to the endoplasmic reticulum (ER) by its signal peptide during synthesis. Proper gp160 folding in the ER requires core glycosylation, disulfide-bond formation and proline isomerization. Signal-peptide cleavage occurs only late after gp160 chain termination and is dependent on folding of the soluble subunit gp120 to a near-native conformation. We here detail the mechanism by which co-translational signal-peptide cleavage is prevented. Conserved residues from the signal peptide and residues downstream of the canonical cleavage site form an extended alpha-helix in the ER membrane, which covers the cleavage site, thus preventing cleavage. A point mutation in the signal peptide breaks the alpha helix allowing co-translational cleavage. We demonstrate that postponed cleavage of gp160 enhances functional folding of the molecule. The change to early cleavage results in decreased viral fitness compared to wild-type HIV.


Subject(s)
HIV Envelope Protein gp160/chemistry , HIV Envelope Protein gp160/metabolism , HIV-1/physiology , Protein Folding , Protein Sorting Signals , Cell Line , Humans , Protein Conformation , Protein Transport , Proteolysis
8.
Chem Commun (Camb) ; 53(13): 2106-2109, 2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28133646

ABSTRACT

Fluorescent proteins (FPs) that can be optically highlighted enable PALM super-resolution microscopy and pulse-chase experiments of cellular molecules. Most FPs evolved in cytoplasmic environments either in the original source organism or in the cytoplasm of bacteria during the course of optimization for research applications. Consequently, many FPs may fold incorrectly in the chemically distinct environments in subcellular organelles. Here, we describe the first monomeric photoswitchable (from green to bright red) FP adapted for oxidizing environments.


Subject(s)
Fluorescent Dyes/analysis , Luminescent Proteins/analysis , Microscopy, Fluorescence/methods , Amino Acid Sequence , Animals , Anthozoa/chemistry , Golgi Apparatus/chemistry , Golgi Apparatus/ultrastructure , HeLa Cells , Humans , Oxidation-Reduction , Photochemical Processes , Sequence Alignment
9.
Mol Ther Nucleic Acids ; 5: e321, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27219515

ABSTRACT

Pathogenic New World hemorrhagic fever mammarenaviruses (NWM) utilize Glycoprotein 1 (GP1) to target the apical domain of the human transferrin receptor (hTfR) for facilitating cell entry. However, the conservation between their GP1s is low. Considering this and the slow evolutionary progression of mammals compared to viruses, therapeutic targeting of hTfR provides an attractive avenue for cross-strain inhibition and diminishing the likelihood of escape mutants. Aptamers present unique advantages for the development of inhibitors to vial entry, including ease of synthesis, lack of immunogenicity, and potentially cold-chain breaking solutions to diseases endemic to South America. Here, recognizing that in vivo competition with the natural ligand, transferrin (Tf), likely drove the evolution of GP1 to recognize the apical domain, we performed competitive in vitro selections against hTfR-expressing cells with supplemented Tf. The resultant minimized aptamer, Waz, binds the apical domain of the receptor and inhibits infection of human cells by recombinant NWM in culture (EC50 ~400 nmol/l). Aptamer multimerization further enhanced inhibition >10-fold (EC50 ~30 nmol/l). Together, our results highlight the ability to use a competitor to bias the outcome of a selection and demonstrate how avidity effects can be leveraged to enhance both aptamer binding and the potency of viral inhibition.

10.
Biotechnol Bioeng ; 113(8): 1647-57, 2016 08.
Article in English | MEDLINE | ID: mdl-26806853

ABSTRACT

We have developed a generalized in vitro compartmentalization-based bead display selection strategy that allows for the identification of enzymes that can perform ligation reactions. Although a number of methods have been developed to evolve such enzymes, many of them are limited in library size (10(6) -10(7) ), do not select for enzymes using a scheme that allows for multiple turnover, or only work on enzymes specific to nucleic acids. This approach is amenable to screening libraries of up to 10(12) protein variants by allowing beads to be overloaded with up to 10(4) unique mutants. Using this approach we isolated a variant of sortase A from Staphylococcus aureus that shows a 114-fold enhancement in kcat /KM in the absence of calcium compared to the wild-type and improved resistance to the inhibitory effects of cell lysates. Unlike the wild-type protein, the newly selected variant shows intracellular activity in the cytoplasm of eukaryotic cells where it may prove useful for intracellular labeling or synthetic biological applications. Biotechnol. Bioeng. 2016;113: 1647-1657. © 2016 Wiley Periodicals, Inc.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/metabolism , Peptide Library , Protein Binding , Protein Engineering/methods , Repressor Proteins/metabolism , Aminoacyltransferases/chemistry , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cloning, Molecular , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Escherichia coli , Kinetics , Models, Molecular , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics
11.
J Biol Chem ; 290(39): 23497-514, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26265468

ABSTRACT

Gap junctions (GJs) are made up of plaques of laterally clustered intercellular channels and the membranes in which the channels are embedded. Arrangement of channels within a plaque determines subcellular distribution of connexin binding partners and sites of intercellular signaling. Here, we report the discovery that some connexin types form plaque structures with strikingly different degrees of fluidity in the arrangement of the GJ channel subcomponents of the GJ plaque. We uncovered this property of GJs by applying fluorescence recovery after photobleaching to GJs formed from connexins fused with fluorescent protein tags. We found that connexin 26 (Cx26) and Cx30 GJs readily diffuse within the plaque structures, whereas Cx43 GJs remain persistently immobile for more than 2 min after bleaching. The cytoplasmic C terminus of Cx43 was required for stability of Cx43 plaque arrangement. We provide evidence that these qualitative differences in GJ arrangement stability reflect endogenous characteristics, with the caveat that the sizes of the GJs examined were necessarily large for these measurements. We also uncovered an unrecognized effect of non-monomerized fluorescent protein on the dynamically arranged GJs and the organization of plaques composed of multiple connexin types. Together, these findings redefine our understanding of the GJ plaque structure and should be considered in future studies using fluorescent protein tags to probe dynamics of highly ordered protein complexes.


Subject(s)
Connexins/metabolism , Gap Junctions/metabolism , Luminescent Proteins/metabolism , Connexin 26 , Connexin 30 , Gap Junctions/chemistry , HeLa Cells , Humans , Protein Conformation
12.
J Virol ; 89(19): 9706-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26202231

ABSTRACT

Many longstanding questions about dynamics of virus-cell interactions can be answered by combining fluorescence imaging techniques with fluorescent protein (FP) tagging strategies. Successfully creating a FP fusion with a cellular or viral protein of interest first requires selecting the appropriate FP. However, while viral architecture and cellular localization often dictate the suitability of a FP, a FP's chemical and physical properties must also be considered. Here, we discuss the challenges of and offer suggestions for identifying the optimal FPs for studying the cell biology of viruses.


Subject(s)
Cellular Microenvironment , Luminescent Proteins/metabolism , Models, Molecular , Optical Imaging/methods , Viral Fusion Proteins/metabolism , Virology/methods , Luminescent Proteins/chemistry , Viral Fusion Proteins/chemistry
13.
Nat Commun ; 6: 7670, 2015 07 09.
Article in English | MEDLINE | ID: mdl-26158227

ABSTRACT

To perform quantitative live cell imaging, investigators require fluorescent reporters that accurately report protein localization and levels, while minimally perturbing the cell. Yet, within the biochemically distinct environments of cellular organelles, popular fluorescent proteins (FPs), including EGFP, can be unreliable for quantitative imaging, resulting in the underestimation of protein levels and incorrect localization. Specifically, within the secretory pathway, significant populations of FPs misfold and fail to fluoresce due to non-native disulphide bond formation. Furthermore, transmembrane FP-fusion constructs can disrupt organelle architecture due to oligomerizing tendencies of numerous common FPs. Here, we describe a powerful set of bright and inert FPs optimized for use in multiple cellular compartments, especially oxidizing environments and biological membranes. Also, we provide new insights into the use of red FPs in the secretory pathway. Our monomeric 'oxFPs' finally resolve long-standing, underappreciated and important problems of cell biology and should be useful for a number of applications.


Subject(s)
Cell Membrane/metabolism , Fluorescent Dyes/metabolism , Luminescent Proteins/metabolism , Animals , Bacterial Proteins/chemistry , Cell Line, Tumor , Dogs , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Luminescent Proteins/chemistry , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence , Optical Imaging/methods , Staining and Labeling , Red Fluorescent Protein
14.
Curr Biol ; 25(10): 1296-305, 2015 May 18.
Article in English | MEDLINE | ID: mdl-25936552

ABSTRACT

Eukaryotic protein secretion requires efficient and accurate delivery of diverse secretory and membrane proteins. This process initiates in the ER, where vesicles are sculpted by the essential COPII coat. The Sec13p subunit of the COPII coat contributes to membrane scaffolding, which enforces curvature on the nascent vesicle. A requirement for Sec13p can be bypassed when traffic of lumenally oriented membrane proteins is abrogated. Here we sought to further explore the impact of cargo proteins on vesicle formation. We show that efficient ER export of the p24 family of proteins is a major driver of the requirement for Sec13p. The scaffolding burden presented by the p24 complex is met in part by the cargo adaptor Lst1p, which binds to a subset of cargo, including the p24 proteins. We propose that the scaffolding function of Lst1p is required to generate vesicles that can accommodate difficult cargo proteins that include large oligomeric assemblies and asymmetrically distributed membrane proteins. Vesicles that contain such cargoes are also more dependent on scaffolding by Sec13p, and may serve as a model for large carrier formation in other systems.


Subject(s)
COP-Coated Vesicles/chemistry , Cell Membrane/metabolism , Membrane Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Molecular Sequence Data , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Transport , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
15.
Virology ; 476: 240-248, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25555152

ABSTRACT

The HIV-1 envelope glycoprotein, gp120, binds the host cell receptor, CD4, in the initial step of HIV viral entry and infection. This process is an appealing target for the development of inhibitory drugs and neutralizing antibodies. To study gp120 binding and intracellular trafficking, we engineered a fluorescent fusion of the humanized gp120 JRFL HIV-1 variant and GFP. Gp120-sfGFP is glycosylated with human sugars, robustly expressed, and secreted from cultured human cells. Protein dynamics, quality control, and trafficking can be visualized in live cells. The fusion protein can be readily modified with different gp120 variants or fluorescent proteins. Finally, secreted gp120-sfGFP enables a sensitive and easy binding assay that can quantitatively screen potential inhibitors of gp120-CD4 binding on live cells via fluorescence imaging or laser scanning cytometry. This adaptable research tool should aid in studies of gp120 cell biology and the development of novel anti-HIV drugs.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Tracking/methods , Green Fluorescent Proteins/metabolism , HIV Envelope Protein gp120/metabolism , HIV Infections/virology , HIV-1/metabolism , CD4-Positive T-Lymphocytes/metabolism , Genetic Engineering , Green Fluorescent Proteins/genetics , HIV Envelope Protein gp120/genetics , HIV Infections/metabolism , HIV-1/genetics , Humans , Receptors, Virus/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Endoplasmic Reticulum Stress Dis ; 1(1): 27-39, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-25419521

ABSTRACT

The endoplasmic reticulum (ER) is the point of entry of proteins into the secretory pathway. Nascent peptides interact with the ER quality control machinery that ensures correct folding of the nascent proteins. Failure to properly fold proteins can lead to loss of protein function and cytotoxic aggregation of misfolded proteins that can lead to cell death. To cope with increases in the ER unfolded secretory protein burden, cells have evolved the Unfolded Protein Response (UPR). The UPR is the primary signaling pathway that monitors the state of the ER folding environment. When the unfolded protein burden overwhelms the capacity of the ER quality control machinery, a state termed ER stress, sensor proteins detect accumulation of misfolded peptides and trigger the UPR transcriptional response. The UPR, which is conserved from yeast to mammals, consists of an ensemble of complex signaling pathways that aims at adapting the ER to the new misfolded protein load. To determine how different factors impact the ER folding environment, various tools and assays have been developed. In this review, we discuss recent advances in live cell imaging reporters and model systems that enable researchers to monitor changes in the unfolded secretory protein burden and activation of the UPR and its associated signaling pathways.

17.
Cell ; 158(3): 534-48, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25018104

ABSTRACT

Depending on endoplasmic reticulum (ER) stress levels, the ER transmembrane multidomain protein IRE1α promotes either adaptation or apoptosis. Unfolded ER proteins cause IRE1α lumenal domain homo-oligomerization, inducing trans autophosphorylation that further drives homo-oligomerization of its cytosolic kinase/endoribonuclease (RNase) domains to activate mRNA splicing of adaptive XBP1 transcription factor. However, under high/chronic ER stress, IRE1α surpasses an oligomerization threshold that expands RNase substrate repertoire to many ER-localized mRNAs, leading to apoptosis. To modulate these effects, we developed ATP-competitive IRE1α Kinase-Inhibiting RNase Attenuators-KIRAs-that allosterically inhibit IRE1α's RNase by breaking oligomers. One optimized KIRA, KIRA6, inhibits IRE1α in vivo and promotes cell survival under ER stress. Intravitreally, KIRA6 preserves photoreceptor functional viability in rat models of ER stress-induced retinal degeneration. Systemically, KIRA6 preserves pancreatic ß cells, increases insulin, and reduces hyperglycemia in Akita diabetic mice. Thus, IRE1α powerfully controls cell fate but can itself be controlled with small molecules to reduce cell degeneration.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Allosteric Regulation , Animals , Apoptosis/drug effects , Cell Line , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Enzyme Activation/drug effects , Humans , Islets of Langerhans/metabolism , Male , Mice , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Rats , Retina/metabolism , Ribonucleases/antagonists & inhibitors
18.
Hum Mutat ; 35(9): 1101-13, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24930953

ABSTRACT

Early-onset dystonia is associated with the deletion of one of a pair of glutamic acid residues (c.904_906delGAG/c.907_909delGAG; p.Glu302del/Glu303del; ΔE 302/303) near the carboxyl-terminus of torsinA, a member of the AAA(+) protein family that localizes to the endoplasmic reticulum lumen and nuclear envelope. This deletion commonly underlies early-onset DYT1 dystonia. While the role of the disease-causing mutation, torsinAΔE, has been established through genetic association studies, it is much less clear whether other rare human variants of torsinA are pathogenic. Two missense variations have been described in single patients: R288Q (c.863G>A; p.Arg288Gln; R288Q) identified in a patient with onset of severe generalized dystonia and myoclonus since infancy and F205I (c.613T>A, p.Phe205Ile; F205I) in a psychiatric patient with late-onset focal dystonia. In this study, we have undertaken a series of analyses comparing the biochemical and cellular effects of these rare variants to torsinAΔE and wild-type (wt) torsinA to reveal whether there are common dysfunctional features. The results revealed that the variants, R288Q and F205I, are more similar in their properties to torsinAΔE protein than to torsinAwt. These findings provide functional evidence for the potential pathogenic nature of these rare sequence variants in the TOR1A gene, thus implicating these pathologies in the development of dystonia.


Subject(s)
Dystonia Musculorum Deformans/genetics , Genetic Variation , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Gene Knockdown Techniques , Genetic Association Studies , Humans , Models, Molecular , Molecular Chaperones/metabolism , Molecular Dynamics Simulation , Mutation , Phenotype , Protein Conformation , Protein Multimerization , Protein Transport , Viral Envelope Proteins/metabolism
19.
Elife ; 3: e01883, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24843009

ABSTRACT

In many cell types, lateral diffusion barriers compartmentalize the plasma membrane and, at least in budding yeast, the endoplasmic reticulum (ER). However, the molecular nature of these barriers, their mode of action and their cellular functions are unclear. Here, we show that misfolded proteins of the ER remain confined into the mother compartment of budding yeast cells. Confinement required the formation of a lateral diffusion barrier in the form of a distinct domain of the ER-membrane at the bud neck, in a septin-, Bud1 GTPase- and sphingolipid-dependent manner. The sphingolipids, but not Bud1, also contributed to barrier formation in the outer membrane of the dividing nucleus. Barrier-dependent confinement of ER stress into the mother cell promoted aging. Together, our data clarify the physical nature of lateral diffusion barriers in the ER and establish the role of such barriers in the asymmetric segregation of proteotoxic misfolded proteins during cell division and aging.DOI: http://dx.doi.org/10.7554/eLife.01883.001.


Subject(s)
Cell Division , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Saccharomyces cerevisiae/metabolism , Sphingolipids/metabolism , Cell Cycle Proteins/metabolism , Diffusion , Guanine Nucleotide Exchange Factors/metabolism , Microfilament Proteins/metabolism , Nuclear Envelope/metabolism , Permeability , Protein Folding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/metabolism , Septins/metabolism , Time Factors , rab GTP-Binding Proteins/metabolism
20.
J Virol ; 88(12): 6922-33, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24696489

ABSTRACT

UNLABELLED: Alphaviruses are small enveloped RNA viruses with highly organized structures that exclude host cell proteins. They contain an internal nucleocapsid and an external lattice of the viral E2 and E1 transmembrane proteins. Alphaviruses bud from the plasma membrane (PM), but the process and dynamics of alphavirus assembly and budding are poorly understood. Here we generated Sindbis viruses (SINVs) with fluorescent protein labels on the E2 envelope protein and exploited them to characterize virus assembly and budding in living cells. During virus infection, E2 became enriched in localized patches on the PM and in filopodium-like extensions. These E2-labeled patches and extensions contained all of the viral structural proteins. Correlative light and electron microscopy studies established that the patches and extensions colocalized with virus budding structures, while light microscopy showed that they excluded a freely diffusing PM marker protein. Exclusion required the interaction of the E2 protein with the capsid protein, a critical step in virus budding, and was associated with the immobilization of the envelope proteins on the cell surface. Virus infection induced two distinct types of extensions: tubulin-negative extensions that were ∼2 to 4 µm in length and excluded the PM marker, and tubulin-positive extensions that were >10 µm long, contained the PM marker, and could transfer virus particles to noninfected cells. Tubulin-positive extensions were selectively reduced in cells infected with a nonbudding SINV mutant. Together, our data support a model in which alphavirus infection induces reorganization of the PM and cytoskeleton, leading to virus budding from specialized sites. IMPORTANCE: Alphaviruses are important and widely distributed human pathogens for which vaccines and antiviral therapies are urgently needed. These small highly organized viruses bud from the host cell PM. Virus assembly and budding are critical but little understood steps in the alphavirus life cycle. We developed alphaviruses with fluorescent protein tags on one of the viral membrane (envelope) proteins and used a variety of microscopy techniques to follow the envelope protein and a host cell PM protein during budding. We showed that alphavirus infection induced the formation of patches and extensions on the PM where the envelope proteins accumulate. These sites excluded other PM proteins and correlated with virus budding structures. Exclusion of PM proteins required specific interactions of the viral envelope proteins with the internal capsid protein. Together, our data indicate that alphaviruses extensively reorganize the cell surface and cytoskeleton to promote their assembly and budding.


Subject(s)
Alphavirus Infections/virology , Sindbis Virus/physiology , Virus Assembly , Virus Release , Animals , Cell Line , Cell Membrane/chemistry , Cell Membrane/virology , Fluorescence Recovery After Photobleaching , Humans , Sindbis Virus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...