Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 23(6): 1871-1882, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38713528

ABSTRACT

The coevolution of liquid chromatography (LC) with mass spectrometry (MS) has shaped contemporary proteomics. LC hyphenated to MS now enables quantification of more than 10,000 proteins in a single injection, a number that likely represents most proteins in specific human cells or tissues. Separations by ion mobility spectrometry (IMS) have recently emerged to complement LC and further improve the depth of proteomics. Given the theoretical advantages in speed and robustness of IMS in comparison to LC, we envision that ongoing improvements to IMS paired with MS may eventually make LC obsolete, especially when combined with targeted or simplified analyses, such as rapid clinical proteomics analysis of defined biomarker panels. In this perspective, we describe the need for faster analysis that might drive this transition, the current state of direct infusion proteomics, and discuss some technical challenges that must be overcome to fully complete the transition to entirely gas phase proteomics.


Subject(s)
Ion Mobility Spectrometry , Proteomics , Proteomics/methods , Ion Mobility Spectrometry/methods , Humans , Chromatography, Liquid/methods , Mass Spectrometry/methods , High-Throughput Screening Assays/methods
2.
bioRxiv ; 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37577577

ABSTRACT

Daptomycin is a last-resort lipopeptide antibiotic that disrupts cell membrane (CM) and peptidoglycan homeostasis. Enterococcus faecalis has developed a sophisticated mechanism to avoid daptomycin killing by re-distributing CM anionic phospholipids away from the septum. The CM changes are orchestrated by a three-component regulatory system, designated LiaFSR, with a possible contribution of cardiolipin synthase (Cls). However, the mechanism by which LiaFSR controls the CM response and the role of Cls are unknown. Here, we show that cardiolipin synthase activity is essential for anionic phospholipid redistribution and daptomycin resistance since deletion of the two genes ( cls1 and cls2 ) encoding Cls abolished CM remodeling. We identified LiaY, a transmembrane protein regulated by LiaFSR, as an important mediator of CM remodeling required for re-distribution of anionic phospholipid microdomains via interactions with Cls1. Together, our insights provide a mechanistic framework on the enterococcal response to cell envelope antibiotics that could be exploited therapeutically.

3.
J Mol Cell Cardiol ; 158: 115-127, 2021 09.
Article in English | MEDLINE | ID: mdl-34081952

ABSTRACT

RATIONALE: The nutrient sensing mechanistic target of rapamycin complex 1 (mTORC1) and its primary inhibitor, tuberin (TSC2), are cues for the development of cardiac hypertrophy. The phenotype of mTORC1 induced hypertrophy is unknown. OBJECTIVE: To examine the impact of sustained mTORC1 activation on metabolism, function, and structure of the adult heart. METHODS AND RESULTS: We developed a mouse model of inducible, cardiac-specific sustained mTORC1 activation (mTORC1iSA) through deletion of Tsc2. Prior to hypertrophy, rates of glucose uptake and oxidation, as well as protein and enzymatic activity of glucose 6-phosphate isomerase (GPI) were decreased, while intracellular levels of glucose 6-phosphate (G6P) were increased. Subsequently, hypertrophy developed. Transcript levels of the fetal gene program and pathways of exercise-induced hypertrophy increased, while hypertrophy did not progress to heart failure. We therefore examined the hearts of wild-type mice subjected to voluntary physical activity and observed early changes in GPI, followed by hypertrophy. Rapamycin prevented these changes in both models. CONCLUSION: Activation of mTORC1 in the adult heart triggers the development of a non-specific form of hypertrophy which is preceded by changes in cardiac glucose metabolism.


Subject(s)
Cardiomegaly/metabolism , Gene Knockdown Techniques/methods , Glucose/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/genetics , Animals , Cardiomegaly/diet therapy , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cells, Cultured , Diet/methods , Disease Models, Animal , Enzyme Activation/genetics , Glucose-6-Phosphatase/metabolism , Isomerases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Phosphorylation/genetics , Sirolimus/administration & dosage , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
4.
Biochemistry ; 59(19): 1854-1868, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32363862

ABSTRACT

The lipid environment in which membrane proteins are embedded can influence their structure and function. Lipid-protein interactions and lipid-induced conformational changes necessary for protein function remain intractable in vivo using high-resolution techniques. Using Escherichia coli strains in which the normal phospholipid composition can be altered or foreign lipids can be introduced, we established the importance of membrane lipid composition for the proper folding, assembly, and function of E. coli lactose (LacY) and sucrose (CscB) permeases. However, the molecular mechanism underlying the lipid dependence for active transport remains unknown. Herein, we demonstrate that the structure and function of CscB and LacY can be modulated by the composition of the lipid environment. Using a combination of assays (transport activity of the substrate, protein topology, folding, and assembly into the membrane), we found that alterations in the membrane lipid composition lead to lipid-dependent structural changes in CscB and LacY. These changes affect the orientation of residues involved in LacY proton translocation and impact the rates of protonation and deprotonation of E325 by affecting the arrangement of transmembrane domains in the vicinity of the R302-E325 charge pair. Furthermore, the structural changes caused by changes in membrane lipid composition can be altered by a single-point mutation, highlighting the adaptability of these transporters to their environment. Altogether, our results demonstrate that direct interactions between a protein and its lipid environment uniquely contribute to membrane protein organization and function. Because members of the major facilitator superfamily present with well-conserved functional architecture, we anticipate that our findings can be extrapolated to other membrane protein transporters.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Membrane Lipids/metabolism , Membrane Transport Proteins/metabolism , Monosaccharide Transport Proteins/metabolism , Symporters/metabolism , Escherichia coli Proteins/chemistry , Membrane Lipids/chemistry , Membrane Transport Proteins/chemistry , Models, Molecular , Monosaccharide Transport Proteins/chemistry , Symporters/chemistry
5.
Sci Rep ; 10(1): 6296, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286407

ABSTRACT

Translocation of preproteins across the Escherichia coli inner membrane requires anionic lipids by virtue of their negative head-group charge either in vivo or in situ. However, available results do not differentiate between the roles of monoanionic phosphatidylglycerol and dianionic cardiolipin (CL) in this essential membrane-related process. To define in vivo the molecular steps affected by the absence of CL in protein translocation and insertion, we analyzed translocon activity, SecYEG stability and its interaction with SecA in an E. coli mutant devoid of CL. Although no growth defects were observed, co- and post-translational translocation of α-helical proteins across inner membrane and the assembly of outer membrane ß-barrel precursors were severely compromised in CL-lacking cells. Components of proton-motive force which could impair protein insertion into and translocation across the inner membrane, were unaffected. However, stability of the dimeric SecYEG complex and oligomerization properties of SecA were strongly compromised while the levels of individual SecYEG translocon components, SecA and insertase YidC were largely unaffected. These results demonstrate that CL is required in vivo for the stability of the bacterial translocon and its efficient function in co-translational insertion into and translocation across the inner membrane of E. coli.


Subject(s)
Cardiolipins/metabolism , Cell Membrane/metabolism , Escherichia coli/metabolism , SEC Translocation Channels/metabolism , Cardiolipins/genetics , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Mutation , Protein Stability , Protein Transport , SecA Proteins/metabolism
6.
J Biol Chem ; 294(49): 18853-18862, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31645436

ABSTRACT

Posttranslational modifications of proteins, such as phosphorylation and dephosphorylation, play critical roles in cellular functions through diverse cell signaling pathways. Protein kinases and phosphatases have been described early on as key regulatory elements of the phosphorylated state of proteins. Tight spatial and temporal regulation of protein kinase and phosphatase activities has to be achieved in the cell to ensure accurate signal transduction. We demonstrated previously that phosphorylation of a membrane protein can lead to its topological rearrangement. Additionally, we found that both the rate and extent of topological rearrangement upon phosphorylation are lipid charge- and lipid environment-dependent. Here, using a model membrane protein (the bacterial lactose permease LacY reconstituted in proteoliposomes) and a combination of real-time measurements and steady-state assessments of protein topology, we established a set of experimental conditions to dissect the effects of phosphorylation and dephosphorylation of a membrane protein on its topological orientation. We also demonstrate that the phosphorylation-induced topological switch of a membrane protein can be reversed upon protein dephosphorylation, revealing a new regulatory role for phosphorylation/dephosphorylation cycles. Furthermore, we determined that the rate of topological rearrangement reversal is correlated with phosphatase activity and is influenced by the membrane's lipid composition, presenting new insights into the spatiotemporal control of the protein phosphorylation state. Together, our results highlight the importance of the compartmentalization of phosphorylation/dephosphorylation cycles in controlling membrane protein topology and, therefore, function, which are influenced by the local lipid environment of the membrane protein.


Subject(s)
Membrane Lipids/chemistry , Membrane Lipids/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Protein Folding , Protein Processing, Post-Translational , Proteolipids/metabolism
7.
Sci Rep ; 9(1): 11338, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31383935

ABSTRACT

Membrane proteins play key roles in cellular functions, their activity mainly depending on their topological arrangement in membranes. Structural studies of membrane proteins have long adopted a protein-centric view regarding the determinants of membrane protein topology and function. Several studies have shown that the orientation of transmembrane domains of polytopic membrane proteins with respect to the plane of the lipid bilayer can be largely determined by membrane lipid composition. However, the mechanism by which membrane proteins exhibit structural and functional duality in the same membrane or different membranes is still unknown. Here we show that lipid-dependent structural and functional assessment of a membrane protein can be conducted in detergent micelles, opening the possibility for the determination of lipid-dependent high-resolution crystal structures. We found that the lactose permease purified from Escherichia coli cells exhibiting varied phospholipid compositions exhibits the same topology and similar function as in its membrane of origin. Furthermore, we found several conditions, including protein mutations and micelle lipid composition, that lead to increased protein stability, correlating with a higher yield of two-dimensional crystal formation. Altogether, our results demonstrate how the membrane lipid environment influences membrane protein topology and arrangement, both in native membranes and in mixed detergent micelles.


Subject(s)
Detergents/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Micelles , Monosaccharide Transport Proteins/chemistry , Phospholipids/chemistry , Symporters/chemistry , Models, Molecular , Protein Conformation , Protein Stability
8.
Protein J ; 38(3): 274-288, 2019 06.
Article in English | MEDLINE | ID: mdl-30937648

ABSTRACT

Due to the heterogenous lipid environment in which integral membrane proteins are embedded, they should follow a set of assembly rules, which govern transmembrane protein folding and topogenesis accordingly to a given lipid profile. Recombinant strains of bacteria have been engineered to have different membrane phospholipid compositions by molecular genetic manipulation of endogenous and foreign genes encoding lipid biosynthetic enzymes. Such strains provide a means to investigate the in vivo role of lipids in many different aspects of membrane function, folding and biogenesis. In vitro and in vivo studies established a function of lipids as molecular chaperones and topological determinants specifically assisting folding and topogenesis of membrane proteins. These results led to the extension of the Positive Inside Rule to Charge Balance Rule, which incorporates a role for lipid-protein interactions in determining membrane protein topological organization at the time of initial membrane insertion and dynamically after initial assembly. Membrane protein topogenesis appears to be a thermodynamically driven process in which lipid-protein interactions affect the potency of charged amino acid residues as topological signals. Dual topology for a membrane protein can be established during initial assembly where folding intermediates in multiple topological conformations are in rapid equilibrium (thus separated by a low activation energy), which is determined by the lipid environment. Post-assembly changes in lipid composition or post-translational modifications can trigger a reorganization of protein topology by inducing destabilization and refolding of a membrane protein. The lipid-dependent dynamic nature of membrane protein organization provides a novel means of regulating protein function.


Subject(s)
Cell Membrane/metabolism , Escherichia coli Proteins , Lipid Bilayers , Membrane Proteins , Phospholipids , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Protein Folding , Protein Processing, Post-Translational
9.
Oncogene ; 38(12): 2135-2150, 2019 03.
Article in English | MEDLINE | ID: mdl-30459358

ABSTRACT

Triple-negative breast cancer (TNBC), the most aggressive breast cancer subtype, currently lacks effective targeted therapy options. Eicosapentaenoic acid (EPA), an omega-3 fatty acid and constituent of fish oil, is a common supplement with anti-inflammatory properties. Although it is not a mainstream treatment, several preclinical studies have demonstrated that EPA exerts anti-tumor activity in breast cancer. However, against solid tumors, EPA as a monotherapy is clinically ineffective; thus, we sought to develop a novel targeted drug combination to bolster its therapeutic action against TNBC. Using a high-throughput functional siRNA screen, we identified Ephrin type-A receptor 2 (EPHA2), an oncogenic cell-surface receptor tyrosine kinase, as a therapeutic target that sensitizes TNBC cells to EPA. EPHA2 expression was uniquely elevated in TNBC cell lines and patient tumors. In independent functional expression studies in TNBC models, EPHA2 gene-silencing combined with EPA significantly reduced cell growth and enhanced apoptosis compared with monotherapies, both in vitro and in vivo. EPHA2-specific inhibitors similarly enhanced the therapeutic action of EPA. Finally, we identified that therapy-mediated apoptosis was attributed to a lethal increase in cancer cell membrane polarity due to ABCA1 inhibition and subsequent dysregulation of cholesterol homeostasis. This study provides new molecular and preclinical evidence to support a clinical evaluation of EPA combined with EPHA2 inhibition in patients with TNBC.


Subject(s)
Cholesterol/metabolism , Eicosapentaenoic Acid/pharmacology , Receptor, EphA2/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , ATP Binding Cassette Transporter 1/metabolism , Animals , Apoptosis/drug effects , Biological Transport/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Drug Interactions , Eicosapentaenoic Acid/therapeutic use , Female , Humans , Mice , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays
10.
Nat Commun ; 8: 15957, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28695917

ABSTRACT

Most bacteria divide using a protein machine called the divisome that spans the cytoplasmic membrane. Key divisome proteins on the membrane's cytoplasmic side include tubulin-like FtsZ, which forms GTP-dependent protofilaments, and actin-like FtsA, which tethers FtsZ to the membrane. Here we present genetic evidence that in Escherichia coli, FtsA antagonizes FtsZ protofilament bundling in vivo. We then show that purified FtsA does not form straight polymers on lipid monolayers as expected, but instead assembles into dodecameric minirings, often in hexameric arrays. When coassembled with FtsZ on lipid monolayers, these FtsA minirings appear to guide FtsZ to form long, often parallel, but unbundled protofilaments, whereas a mutant of FtsZ (FtsZ*) with stronger lateral interactions remains bundled. In contrast, a hypermorphic mutant of FtsA (FtsA*) forms mainly arcs instead of minirings and enhances lateral interactions between FtsZ protofilaments. Based on these results, we propose that FtsA antagonizes lateral interactions between FtsZ protofilaments, and that the oligomeric state of FtsA may influence FtsZ higher-order structure and divisome function.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Lipid Bilayers/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Lipid Bilayers/chemistry , Lipids/chemistry , Protein Binding
11.
J Bacteriol ; 199(13)2017 07 01.
Article in English | MEDLINE | ID: mdl-28439040

ABSTRACT

Bacteria have evolved multiple strategies to sense and rapidly adapt to challenging and ever-changing environmental conditions. The ability to alter membrane lipid composition, a key component of the cellular envelope, is crucial for bacterial survival and adaptation in response to environmental stress. However, the precise roles played by membrane phospholipids in bacterial physiology and stress adaptation are not fully elucidated. The goal of this study was to define the role of membrane phospholipids in adaptation to stress and maintenance of bacterial cell fitness. By using genetically modified strains in which the membrane phospholipid composition can be systematically manipulated, we show that alterations in major Escherichia coli phospholipids transform these cells globally. We found that alterations in phospholipids impair the cellular envelope structure and function, the ability to form biofilms, and bacterial fitness and cause phospholipid-dependent susceptibility to environmental stresses. This study provides an unprecedented view of the structural, signaling, and metabolic pathways in which bacterial phospholipids participate, allowing the design of new approaches in the investigation of lipid-dependent processes involved in bacterial physiology and adaptation.IMPORTANCE In order to cope with and adapt to a wide range of environmental conditions, bacteria have to sense and quickly respond to fluctuating conditions. In this study, we investigated the effects of systematic and controlled alterations in bacterial phospholipids on cell shape, physiology, and stress adaptation. We provide new evidence that alterations of specific phospholipids in Escherichia coli have detrimental effects on cellular shape, envelope integrity, and cell physiology that impair biofilm formation, cellular envelope remodeling, and adaptability to environmental stresses. These findings hold promise for future antibacterial therapies that target bacterial lipid biosynthesis.


Subject(s)
Cell Membrane/physiology , Escherichia coli/physiology , Phospholipids/metabolism , Bacterial Adhesion , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Biofilms , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli/ultrastructure , Gene Expression Regulation, Bacterial/physiology , Homeostasis/physiology , Lipopolysaccharides/metabolism , Stress, Physiological
12.
Biochim Biophys Acta Biomembr ; 1859(7): 1291-1300, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28432030

ABSTRACT

The final topology of membrane proteins is thought to be dictated primarily by the encoding sequence. However, according to the Charge Balance Rule the topogenic signals within nascent membrane proteins are interpreted in agreement with the Positive Inside Rule as influenced by the protein phospholipid environment. The role of long-range protein-lipid interactions in establishing a final uniform or dual topology is unknown. In order to address this role, we determined the positional dependence of the potency of charged residues as topological signals within Escherichia coli sucrose permease (CscB) in cells in which the zwitterionic phospholipid phosphatidylethanolamine (PE), acting as topological determinant, was either eliminated or tightly titrated. Although the position of a single or paired oppositely charged amino acid residues within an extramembrane domain (EMD), either proximal, central or distal to a transmembrane domain (TMD) end, does not appear to be important, the oppositely charged residues exert their topogenic effects separately only in the absence of PE. Thus, the Charge Balance Rule can be executed in a retrograde manner from any cytoplasmic EMD or any residue within an EMD most likely outside of the translocon. Moreover, CscB is inserted into the membrane in two opposite orientations at different ratios with the native orientation proportional to the mol % of PE. The results demonstrate how the cooperative contribution of lipid-protein interactions affects the potency of charged residues as topological signals, providing a molecular mechanism for the realization of single, equal or different amounts of oppositely oriented protein within the same membrane.


Subject(s)
Escherichia coli Proteins/metabolism , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Amino Acid Sequence , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry
13.
J Biol Chem ; 292(5): 1613-1624, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27974465

ABSTRACT

Membrane protein topology and folding are governed by structural principles and topogenic signals that are recognized and decoded by the protein insertion and translocation machineries at the time of initial membrane insertion and folding. We previously demonstrated that the lipid environment is also a determinant of initial protein topology, which is dynamically responsive to post-assembly changes in membrane lipid composition. However, the effect on protein topology of post-assembly phosphorylation of amino acids localized within initially cytoplasmically oriented extramembrane domains has never been investigated. Here, we show in a controlled in vitro system that phosphorylation of a membrane protein can trigger a change in topological arrangement. The rate of change occurred on a scale of seconds, comparable with the rates observed upon changes in the protein lipid environment. The rate and extent of topological rearrangement were dependent on the charges of extramembrane domains and the lipid bilayer surface. Using model membranes mimicking the lipid compositions of eukaryotic organelles, we determined that anionic lipids, cholesterol, sphingomyelin, and membrane fluidity play critical roles in these processes. Our results demonstrate how post-translational modifications may influence membrane protein topology in a lipid-dependent manner, both along the organelle trafficking pathway and at their final destination. The results provide further evidence that membrane protein topology is dynamic, integrating for the first time the effect of changes in lipid composition and regulators of cellular processes. The discovery of a new topology regulatory mechanism opens additional avenues for understanding unexplored structure-function relationships and the development of optimized topology prediction tools.


Subject(s)
Cell Membrane/chemistry , Lipid Bilayers/chemistry , Membrane Lipids/chemistry , Membrane Proteins/chemistry , Protein Processing, Post-Translational , Cell Membrane/metabolism , Lipid Bilayers/metabolism , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Phosphorylation , Protein Domains
14.
Proc Natl Acad Sci U S A ; 113(37): 10436-41, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27582470

ABSTRACT

Hematologic malignancies are frequently associated with cardiac pathologies. Mutations of isocitrate dehydrogenase 1 and 2 (IDH1/2) occur in a subset of acute myeloid leukemia patients, causing metabolic and epigenetic derangements. We have now discovered that altered metabolism in leukemic cells has a profound effect on cardiac metabolism. Combining mathematical modeling and in vivo as well as ex vivo studies, we found that increased amounts of the oncometabolite d-2-hydroxyglutarate (D2-HG), produced by IDH2 mutant leukemic cells, cause contractile dysfunction in the heart. This contractile dysfunction is associated with impaired oxidative decarboxylation of α-ketoglutarate, a redirection of Krebs cycle intermediates, and increased ATP citrate lyase (ACL) activity. Increased availability of D2-HG also leads to altered histone methylation and acetylation in the heart. We propose that D2-HG promotes cardiac dysfunction by impairing α-ketoglutarate dehydrogenase and induces histone modifications in an ACL-dependent manner. Collectively, our results highlight the impact of cancer cell metabolism on function and metabolism of the heart.


Subject(s)
ATP Citrate (pro-S)-Lyase/genetics , Cardiomyopathies/genetics , Hematologic Neoplasms/genetics , Isocitrate Dehydrogenase/genetics , Ketoglutarate Dehydrogenase Complex/genetics , Myocardium/metabolism , ATP Citrate (pro-S)-Lyase/metabolism , Acetylation , Animals , Cardiomyopathies/complications , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Glutarates/metabolism , Hematologic Neoplasms/complications , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Histones/genetics , Histones/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Methylation , Mice , Mutation , Myocardium/pathology
15.
Proc Natl Acad Sci U S A ; 112(45): 13874-9, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26512118

ABSTRACT

A fundamental objective in membrane biology is to understand and predict how a protein sequence folds and orients in a lipid bilayer. Establishing the principles governing membrane protein folding is central to understanding the molecular basis for membrane proteins that display multiple topologies, the intrinsic dynamic organization of membrane proteins, and membrane protein conformational disorders resulting in disease. We previously established that lactose permease of Escherichia coli displays a mixture of topological conformations and undergoes postassembly bidirectional changes in orientation within the lipid bilayer triggered by a change in membrane phosphatidylethanolamine content, both in vivo and in vitro. However, the physiological implications and mechanism of dynamic structural reorganization of membrane proteins due to changes in lipid environment are limited by the lack of approaches addressing the kinetic parameters of transmembrane protein flipping. In this study, real-time fluorescence spectroscopy was used to determine the rates of protein flipping in the lipid bilayer in both directions and transbilayer flipping of lipids triggered by a change in proteoliposome lipid composition. Our results provide, for the first time to our knowledge, a dynamic picture of these events and demonstrate that membrane protein topological rearrangements in response to lipid modulations occur rapidly following a threshold change in proteoliposome lipid composition. Protein flipping was not accompanied by extensive lipid-dependent unfolding of transmembrane domains. Establishment of lipid bilayer asymmetry was not required but may accelerate the rate of protein flipping. Membrane protein flipping was found to accelerate the rate of transbilayer flipping of lipids.


Subject(s)
Membrane Proteins/chemistry , Phospholipids/chemistry , Escherichia coli/enzymology , Fluorescence Resonance Energy Transfer , Lipid Bilayers , Membrane Transport Proteins/chemistry , Proteolipids , Spectrometry, Fluorescence
16.
Structure ; 23(4): 612-4, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25862933

ABSTRACT

In this issue of Structure, Serdiuk et al. report the use of single-molecule force microscopy to establish a role for phosphatidylethanolamine in promoting the native fold of lactose permease, thereby preventing it from populating a functionally defective, nonnative conformation (Serdiuk et al., 2015).


Subject(s)
Escherichia coli Proteins/chemistry , Membrane Lipids/chemistry , Molecular Dynamics Simulation , Monosaccharide Transport Proteins/chemistry , Symporters/chemistry
17.
Biochim Biophys Acta ; 1843(8): 1475-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24341994

ABSTRACT

Membrane protein folding and topogenesis are tuned to a given lipid profile since lipids and proteins have co-evolved to follow a set of interdependent rules governing final protein topological organization. Transmembrane domain (TMD) topology is determined via a dynamic process in which topogenic signals in the nascent protein are recognized and interpreted initially by the translocon followed by a given lipid profile in accordance with the Positive Inside Rule. The net zero charged phospholipid phosphatidylethanolamine and other neutral lipids dampen the translocation potential of negatively charged residues in favor of the cytoplasmic retention potential of positively charged residues (Charge Balance Rule). This explains why positively charged residues are more potent topological signals than negatively charged residues. Dynamic changes in orientation of TMDs during or after membrane insertion are attributed to non-sequential cooperative and collective lipid-protein charge interactions as well as long-term interactions within a protein. The proportion of dual topological conformers of a membrane protein varies in a dose responsive manner with changes in the membrane lipid composition not only in vivo but also in vitro and therefore is determined by the membrane lipid composition. Switching between two opposite TMD topologies can occur in either direction in vivo and also in liposomes (designated as fliposomes) independent of any other cellular factors. Such lipid-dependent post-insertional reversibility of TMD orientation indicates a thermodynamically driven process that can occur at any time and in any cell membrane driven by changes in the lipid composition. This dynamic view of protein topological organization influenced by the lipid environment reveals previously unrecognized possibilities for cellular regulation and understanding of disease states resulting from mis-folded proteins. This article is part of a Special Issue entitled: Protein trafficking and secretion in bacteria. Guest Editors: Anastassios Economou and Ross Dalbey.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Phosphatidylethanolamines/metabolism , Protein Transport/genetics , Bacteria/chemistry , Bacteria/metabolism , Cytoplasm/metabolism , Lipids/chemistry , Lipids/genetics , Membrane Proteins/chemistry , Phosphatidylethanolamines/genetics , Protein Folding , Protein Structure, Tertiary/genetics
18.
Proc Natl Acad Sci U S A ; 110(23): 9338-43, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23690595

ABSTRACT

Phospholipids could exert their effect on membrane protein topology either directly by interacting with topogenic signals of newly inserted proteins or indirectly by influencing the protein assembly machinery. In vivo lactose permease (LacY) of Escherichia coli displays a mixture of topological conformations ranging from complete inversion of the N-terminal helical bundle to mixed topology and then to completely native topology as phosphatidylethanolamine (PE) is increased from 0% to 70% of membrane phospholipids. These topological conformers are interconvertible by postassembly synthesis or dilution of PE in vivo. To investigate whether coexistence of multiple topological conformers is dependent solely on the membrane lipid composition, we determined the topological organization of LacY in an in vitro proteoliposome system in which lipid composition can be systematically controlled before (liposomes) and after (fliposomes) reconstitution using a lipid exchange technique. Purified LacY reconstituted into preformed liposomes of increasing PE content displayed inverted topology at low PE and then a mixture of inverted and proper topologies with the latter increasing with increasing PE until all LacY adopted its native topology. Interconversion between topological conformers of LacY was observed in a PE dose-dependent manner by either increasing or decreasing PE levels in proteoliposomes postreconstitution of LacY, clearly demonstrating that membrane protein topology can be changed simply by changing membrane lipid composition independent of other cellular factors. The results provide a thermodynamic-based lipid-dependent model for shifting the equilibrium between different conformational states of a membrane protein.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Monosaccharide Transport Proteins/chemistry , Protein Conformation , Symporters/chemistry , Blotting, Western , Chromatography, Thin Layer , Electrophoresis, Polyacrylamide Gel , Escherichia coli Proteins/genetics , Immunoprecipitation , Liposomes/metabolism , Liposomes/ultrastructure , Microscopy, Electron , Monosaccharide Transport Proteins/genetics , Phospholipids/metabolism , Symporters/genetics
19.
J Biol Chem ; 288(8): 5873-85, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23322771

ABSTRACT

Energy-dependent uphill transport but not energy-independent downhill transport by lactose permease (LacY) is impaired when expressed in Escherichia coli cells or reconstituted in liposomes lacking phosphatidylethanolamine (PE) and containing only anionic phospholipids. The absence of PE results in inversion of the N-terminal half and misfolding of periplasmic domain P7, which are required for uphill transport of substrates. Replacement of PE in vitro by lipids with no net charge (phosphatidylcholine (PC), monoglucosyl diacylglycerol (GlcDAG), or diglucosyl diacylglycerol (GlcGlcDAG)) supported wild type transmembrane topology of the N-terminal half of LacY. The restoration of uphill transport in vitro was dependent on LacY native topology and proper folding of P7. Support of uphill transport by net neutral lipids in vitro (PE > PC ≫ GlcDAG ≠ GlcGlcDAG provided that PE or PC contained one saturated fatty acid) paralleled the results observed previously in vivo (PE = PC > GlcDAG ≠ GlcGlcDAG). Therefore, a free amino group is not required for uphill transport as previously concluded based on the lack of in vitro uphill transport when fully unsaturated PC replaced E. coli-derived PE. A close correlation was observed in vivo and in vitro between the ability of LacY to carry out uphill transport, the native conformation of P7, and the lipid headgroup and fatty acid composition. Therefore, the headgroup and the fatty acid composition of lipids are important for defining LacY topological organization and catalytically important structural features, further illustrating the direct role of lipids, independent of other cellular factors, in defining membrane protein structure/function.


Subject(s)
Amines/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Fatty Acids/chemistry , Lipids/chemistry , Membrane Transport Proteins/chemistry , Monosaccharide Transport Proteins/chemistry , Symporters/chemistry , Biological Transport , Catalysis , Cell Membrane/metabolism , Densitometry/methods , Epitopes/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Monosaccharide Transport Proteins/metabolism , Phosphatidylethanolamines/chemistry , Protein Binding , Proteolipids/metabolism , Symporters/metabolism
20.
J Mol Biol ; 415(3): 584-99, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22119486

ABSTRACT

The accumulation of amyloid fibers due to protein misfolding is associated with numerous human diseases. For example, the formation of amyloid deposits in neurodegenerative pathologies is correlated with abnormal apoptosis. We report here the in vitro formation of various types of aggregates by Bcl-xL, a protein of the Bcl-2 family involved in the regulation of apoptosis. Bcl-xL forms aggregates in three states, micelles, native-like fibrils, and amyloid fibers, and their biophysical characterization has been performed in detail. Bcl-xL remains in its native state within micelles and native-like fibrils, and our results suggest that native-like fibrils are formed by the association of micelles. Formation of amyloid structures, that is, nonnative intermolecular ß-sheets, is favored by the proximity of proteins within fibrils at the expense of the Bcl-xL native structure. Finally, we provide evidence of a direct relationship between the amyloid character of the fibers and the tertiary-structure stability of the native Bcl-xL. The potential causality between the accumulation of Bcl-xL into amyloid deposits and abnormal apoptosis during neurodegenerative diseases is discussed.


Subject(s)
Amyloid/metabolism , bcl-X Protein/metabolism , Amyloid/chemistry , Amyloid/ultrastructure , Humans , Microscopy, Electron , Models, Molecular , Protein Conformation , Protein Denaturation , Protein Multimerization , Protein Stability , bcl-X Protein/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...