Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Heidi Vitrac is active.

Publication


Featured researches published by Heidi Vitrac.


Journal of Biological Chemistry | 2009

Characterization of the Structure and Intermolecular Interactions between the Connexin40 and Connexin43 Carboxyl-terminal and Cytoplasmic Loop Domains

Denis Bouvier; Gaelle Spagnol; Sylvie Chenavas; Fabien Kieken; Heidi Vitrac; Sarah Brownell; Admir Kellezi; Vincent Forge; Paul L. Sorgen

Gap junctions are intercellular channels that allow the passage of ions, small molecules, and second messengers that are essential for the coordination of cellular function. They are formed by two hemichannels, each constituted by the oligomerization of six connexins (Cx). Among the 21 different human Cx isoforms, studies have suggested that in the heart, Cx40 and Cx43 can oligomerize to form heteromeric hemichannels. The mechanism of heteromeric channel regulation has not been clearly defined. Tissue ischemia leads to intracellular acidification and closure of Cx43 and Cx40 homomeric channels. However, coexpression of Cx40 and Cx43 in Xenopus oocytes enhances the pH sensitivity of the channel. This phenomenon requires the carboxyl-terminal (CT) part of both connexins. In this study we used different biophysical methods to determine the structure of the Cx40CT and characterize the Cx40CT/Cx43CT interaction. Our results revealed that the Cx40CT is an intrinsically disordered protein similar to the Cx43CT and that the Cx40CT and Cx43CT can interact. Additionally, we have identified an interaction between the Cx40CT and the cytoplasmic loop of Cx40 as well as between the Cx40CT and the cytoplasmic loop of Cx43 (and vice versa). Our studies support the “particle-receptor” model for pH gating of Cx40 and Cx43 gap junction channels and suggest that interactions between cytoplasmic regulatory domains (both homo- and hetero-connexin) could be important for the regulation of heteromeric channels.


Journal of Biological Chemistry | 2011

Lipids and Topological Rules of Membrane Protein Assembly: BALANCE BETWEEN LONG AND SHORT RANGE LIPID-PROTEIN INTERACTIONS*

Heidi Vitrac; Mikhail Bogdanov; Philip Heacock; William Dowhan

The N-terminal six-transmembrane domain (TM) bundle of lactose permease of Escherichia coli is uniformly inverted when assembled in membranes lacking phosphatidylethanolamine (PE). Inversion is dependent on the net charge of cytoplasmically exposed protein domains containing positive and negative residues, net charge of the membrane surface, and low hydrophobicity of TM VII acting as a molecular hinge between the two halves of lactose permease (Bogdanov, M., Xie, J., Heacock, P., and Dowhan, W. (2008) J. Cell Biol. 182, 925–935). Net neutral lipids suppress the membrane translocation potential of negatively charged amino acids, thus increasing the cytoplasmic retention potential of positively charged amino acids. Herein, TM organization of sucrose permease (CscB) and phenylalanine permease (PheP) as a function of membrane lipid composition was investigated to extend these principles to other proteins. For CscB, topological dependence on PE only becomes evident after a significant increase in the net negative charge of the cytoplasmic surface of the N-terminal TM bundle. High negative charge is required to overcome the thermodynamic block to inversion due to the high hydrophobicity of TM VII. Increasing the positive charge of the cytoplasmic surface of the N-terminal TM hairpin of PheP, which is misoriented in PE-lacking cells, favors native orientation in the absence of PE. PheP and CscB also display co-existing dual topologies dependent on changes in the charge balance between protein domains and the membrane lipids. Therefore, the topology of both permeases is dependent on PE. However, CscB topology is governed by thermodynamic balance between opposing lipid-dependent electrostatic and hydrophobic interactions.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Oncometabolite d-2-hydroxyglutarate impairs α-ketoglutarate dehydrogenase and contractile function in rodent heart.

Anja Karlstaedt; Xiaotian Zhang; Heidi Vitrac; Romain Harmancey; Hernan Vasquez; Jing Han Wang; Margaret A. Goodell; Heinrich Taegtmeyer

Significance We show that the oncometabolite d-2-hydroxyglutarate (D2-HG) affects cardiac function in the isolated working heart by inhibiting α-KGDH, a key regulatory enzyme of cellular energy metabolism. Analyzing metabolic flux rates by using in vitro and ex vivo approaches in combination with integrative mathematical modeling enabled us to identify the mechanisms by which D2-HG perturbs metabolic flux and induces epigenetic modifications in the heart. The results provide knowledge about malignancy-related changes in enzymatic activity and posttranslational modifications in the context of cardiac remodeling. 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.


Journal of Bacteriology | 2017

Impact of membrane phospholipid alterations in Escherichia coli on cellular function and bacterial stress adaptation

Veronica W. Rowlett; Anja Karlstaedt; William Dowhan; Heinrich Taegtmeyer; William Margolin; Heidi Vitrac

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.


Nature Communications | 2017

Escherichia coli FtsA forms lipid-bound minirings that antagonize lateral interactions between FtsZ protofilaments

Marcin Krupka; Veronica W. Rowlett; Dustin R. Morado; Heidi Vitrac; Kara M. Schoenemann; Jun Liu; William Margolin

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.


Journal of Biological Chemistry | 2017

Dynamic Lipid-dependent Modulation of Protein Topology by Post-translational Phosphorylation

Heidi Vitrac; David M. MacLean; Anja Karlstaedt; Heinrich Taegtmeyer; Vasanthi Jayaraman; Mikhail Bogdanov; William Dowhan

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.


Biochimica et Biophysica Acta | 2017

Effects of mixed proximal and distal topogenic signals on the topological sensitivity of a membrane protein to the lipid environment

Heidi Vitrac; William Dowhan; Mikhail Bogdanov

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.


Cancer Research | 2017

Abstract 1235: EPHA2-targeted therapy enhances the cytotoxicity of eicosapentaenoic acid against triple-negative inflammatory breast cancer

Angie M. Torres-Adorno; Heidi Vitrac; Yuan Qi; Yiwen Yang; Peiying Yang; Bedrich L. Eckhardt; Naoto T. Ueno

Background: Inflammatory breast cancer (IBC) is the most aggressive form of breast cancer. We have previously reported that mediators of inflammation, such as COX-2, promote the growth of Triple-Negative receptor (TN) IBC xenografts; therefore, inflammation in TN-IBC has a unique opportunity as a therapeutic strategy. Eicosapentaenoic acid (EPA), a non-toxic omega-3 fatty acid with anti-inflammatory properties, has partially reduced tumor growth in pre-clinical models of TN-IBC. Therefore, our goal is to develop a novel non-toxic approach that enhances EPA efficacy against TN-IBC in combination with targeted therapy. Methods and Results: Using a high-throughput, siRNA screen (939 genes) in the TN-IBC cell line SUM149PT, we identified Ephrin type-A receptor 2 (EPHA2), an oncogenic cell-surface receptor tyrosine kinase, as a target that modulates the sensitivity of TN-IBC cells to EPA treatment. To determine the clinical relevance of EPHA2, we interrogated a meta-analysis of breast cancer mRNA expression data sets, and found that high EPHA2 tumor expression was significantly correlated with poor overall survival in TN-IBC patients, compared to low EPHA2 expressing tumors (P = 0.01). We observed no significant correlations to other breast cancer subtypes. Similar findings were observed in vitro were EPHA2 expression predominantly occurred in the TN-IBC subtypes (19 of 30) among 49 breast cancer cell lines. Gain/loss-of-expression studies were performed to functionally validate EPHA2 as a synergistic combinational target with EPA in two EPHA2-expressing TN-IBC models, SUM149PT and BCX010, using proliferation and apoptosis assays in vitro and established tumor xenografts in vivo. EPHA2 gene silencing significantly reduced cell growth and induced apoptosis in combination with EPA when compared with untreated control and monotherapy in vitro (P Conclusions: Our preclinical findings provide a rationale for the development of a phase 1 clinical trial investigating combination EPA and EPHA2-inhibitors in patients with EPHA2-positive TN-IBC. Citation Format: Angie M. Torres-Adorno, Heidi Vitrac, Yuan Qi, Yiwen Yang, Peiying Yang, Bedrich L. Eckhardt, Naoto T. Ueno. EPHA2-targeted therapy enhances the cytotoxicity of eicosapentaenoic acid against triple-negative inflammatory breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1235. doi:10.1158/1538-7445.AM2017-1235


Biophysical Journal | 2011

Lipids and Topological “rules” of Membrane Protein Assembly: Testing the Generality of Net Charge Balance Rule

Heidi Vitrac; Mikhail Bogdanov; Philip Heacock; William Dowhan

Transmembrane domain (TMD) orientation within some membrane proteins is dependent on membrane lipid composition. When the lactose permease (LacY) is assembled in Escherichia coli membranes lacking the major phospholipid phosphatidylethanolamine (PE), the N-terminal TMD bundle is inverted with respect to the C-terminal TMD bundle and the plane of the membrane. This inversion is dependent on the interfacial net positive charge of the protein, the net negative charge of the membrane and a TMD of low hydrophobicity, acting as a molecular hinge between the two halves of the protein by exiting the membrane to the periplasm. Homologous E. coli sucrose permease (CscB) and non-homologous phenylalanine permease (PheP) were investigated to generalize these original observations.CscB function, like that of LacY, is dependent on the presence of PE but topological dependence on membrane lipid composition is less sensitive and only becomes evident after significant changes in the net positive charge of the cytoplasmic surface of the N-terminal bundle. The first cytoplasmic domains of PheP, which are misoriented in PE-lacking cells, have a net negative charge. Decreasing the negative charge density of the extramembrane domains flanking the N-terminal TMD hairpin of PheP favors a reorientation of the N-terminus and adjacent hairpin to their native orientation in the absence of PE as predicted by the above net charge balance rule.Polytopic membrane proteins containing competing opposite charges within their cytoplasmic domains may share a common mechanism for topogenesis dependent on PE. However the degree of sensitivity to phospholipid composition appears to be sequence-specific and might be a result of conformational flexibility, topological preference of individual domains or the availability of mechanical hinge region. Supported by NIH grant R37-GM20478.


Biophysical Journal | 2018

Lipids as Determinants of Membrane Protein Structure

William Dowhan; Mikhail Bogdanov; Heidi Vitrac

Collaboration


Dive into the Heidi Vitrac's collaboration.

Top Co-Authors

Avatar

William Dowhan

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Mikhail Bogdanov

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Anja Karlstaedt

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Heinrich Taegtmeyer

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Hernan Vasquez

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip Heacock

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Veronica W. Rowlett

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

William Margolin

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Vincent Forge

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge