19 research outputs found

    Diminished Self-Chaperoning Activity of the ΔF508 Mutant of CFTR Results in Protein Misfolding

    Get PDF
    The absence of a functional ATP Binding Cassette (ABC) protein called the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) from apical membranes of epithelial cells is responsible for cystic fibrosis (CF). Over 90% of CF patients carry at least one mutant allele with deletion of phenylalanine at position 508 located in the N-terminal nucleotide binding domain (NBD1). Biochemical and cell biological studies show that the ΔF508 mutant exhibits inefficient biosynthetic maturation and susceptibility to degradation probably due to misfolding of NBD1 and the resultant misassembly of other domains. However, little is known about the direct effect of the Phe508 deletion on the NBD1 folding, which is essential for rational design strategies of cystic fibrosis treatment. Here we show that the deletion of Phe508 alters the folding dynamics and kinetics of NBD1, thus possibly affecting the assembly of the complete CFTR. Using molecular dynamics simulations, we find that meta-stable intermediate states appearing on wild type and mutant folding pathways are populated differently and that their kinetic accessibilities are distinct. The structural basis of the increased misfolding propensity of the ΔF508 NBD1 mutant is the perturbation of interactions in residue pairs Q493/P574 and F575/F578 found in loop S7-H6. As a proof-of-principle that the S7-H6 loop conformation can modulate the folding kinetics of NBD1, we virtually design rescue mutations in the identified critical interactions to force the S7-H6 loop into the wild type conformation. Two redesigned NBD1-ΔF508 variants exhibited significantly higher folding probabilities than the original NBD1-ΔF508, thereby partially rescuing folding ability of the NBD1-ΔF508 mutant. We propose that these observed defects in folding kinetics of mutant NBD1 may also be modulated by structures separate from the 508 site. The identified structural determinants of increased misfolding propensity of NBD1-ΔF508 are essential information in correcting this pathogenic mutant

    Regulatory Insertion Removal Restores Maturation, Stability and Function of ΔF508 CFTR

    Get PDF
    The cystic fibrosis transmembrane conductance regulator (CFTR) epithelial anion channel is a large multi-domain membrane protein which matures inefficiently during biosynthesis. Its assembly is further perturbed by the deletion of F508 from the first nucleotide binding domain (NBD1) responsible for most cystic fibrosis. The mutant polypeptide is recognized by cellular quality control systems and is proteolyzed. CFTR NBD1 contains a 32 residue segment termed the regulatory insertion (RI) not present in other ABC transporters. We report here that RI deletion enabled ΔF508 CFTR to mature and traffic to the cell surface where it mediated regulated anion efflux and exhibited robust single chloride channel activity. Long term pulse-chase experiments showed that the mature ΔRI/ΔF508 had a T1/2 of ~14h in cells, similar to the wild-type. RI deletion restored ATP occlusion by NBD1 of ΔF508 CFTR and had a strong thermo-stabilizing influence on the channel with gating up to at least 40°C. None of these effects of RI removal were achieved by deletion of only portions of RI. Discrete molecular dynamics simulations of NBD1 indicated that RI might indirectly influence the interaction of NBD1 with the rest of the protein by attenuating the coupling of the F508 containing loop with the F1-like ATP-binding core subdomain so that RI removal overcame the perturbations caused by F508 deletion. Restriction of RI to a particular conformational state may ameliorate the impact of the disease-causing mutation

    Investigating the impact of small molecule ligands and the proteostasis network on protein folding inside the cell

    Get PDF
    The folded forms of most proteins are critical to their functions. Despite the complexity of the cellular milieu and the presence of high-risk deleterious interactions, there is a high level of fidelity observed in the folding process for entire proteomes. Two important reasons for this are the presence of the quality control machinery consisting of chaperones and degradation enzymes that work jointly to optimize the population of the folded state and interaction partners that re-enforce the functional state and add to the competitive advantage of an organism. While substantial effort has been directed to understand protein folding and interactions in vitro, comparatively little of these processes are explored inside the cell. This work examines two important aspects of protein folding inside the cell; first, the impact of small molecule ligands on protein folding; and second, the impact of the proteostasis network on the folding of an obligatory chaperone client. We deploy a combination of experiments and mathematical modeling based on the principle of kinetic partitioning to understand how these phenomena sculpt the protein folding landscape inside the cell. We find that ligands specifically deplete unfolded and aggregation- or degradation - prone protein populations by favoring the folded state and the chaperone and degradation proteins work to minimize off-pathway species thus reducing the population of aggregated protein inside the cell

    Structural and Biochemical Studies of Membrane Proteins CFTR and GLUT1 Yield New Insights into the Molecular Basis of Cystic Fibrosis and Biology of Glucose Transport

    Get PDF
    Integral membrane proteins (IMPs) assume critical roles in cell biology and are key targets for drug discovery. Given their involvement in a wide range of diseases, the structural and functional characterization of IMPs are of significant importance. However, this remains notoriously challenging due to the difficulties of stably purifying membrane-bound, hydrophobic proteins. Compounding this, many diseases are caused by IMP mutations that further decrease their stability. One such example is cystic fibrosis (CF), which is caused by misfolding or dysfunction of the epithelial cell chloride channel cystic fibrosis transmembrane conductance regulator (CFTR). Roughly 70% of CF patients world-wide harbor the ΔF508-CFTR mutation, which interrupts CFTR’s folding, maturation, trafficking and function. No existing treatment sufficiently addresses the consequences of ΔF508, and the substantial instability that results from this mutation limits our ability to study ΔF508-CFTR in search of better treatments. To that end, my colleagues at Sanofi generated homology models of full-length wild-type and ΔF508-CFTR +/- second-site suppressor mutations (SSSMs) V510D and R1070W, and performed molecular dynamics (MD) simulations for each model. Using information obtained from this analysis, I tested several hypotheses on the mechanism by which ΔF508 destabilizes full-length CFTR and how SSSMs suppress this effect. Leveraging studies of the purified NBD1 subdomain and of full-length CFTR in a cellular context, I confirmed the prediction of a key salt-bridge interaction between V510D and K564 important to second-site suppression. Furthermore, I identified a novel class of SSSMs that support a key prediction from these analyses: that helical unraveling of TM10, within CFTR’s second transmembrane domain, is an important contributor to ΔF508-induced instability. In addition, I developed a detergent-free CFTR purification method using styrene-maleic acid (SMA) copolymer to extract the channel directly from its cell membrane along with the surrounding lipid content. The resulting particles were stable, monodisperse discs containing a single molecule of highly-purified CFTR. With this material, I optimized grid preparation techniques and carried out cryo-EM structural analysis of WT-hCFTR which resulted in 2D particle class averages which were consistent with an ABC transporter shape characteristic of CFTR, and a preliminary 3D reconstruction. This result establishes a foundation for future characterization of ΔF508-CFTR in its native state. I have also applied this SMA-based purification method to the facilitated glucose transporter GLUT1 (SLC2A1). SLC2A1 mutations contribute to a rare and developmentally debilitating disease called GLUT1-deficiency syndrome. Using SMA, I successfully extracted GLUT1 in its native state. With the application of this method, I was able to purify endogenous GLUT1 from erythrocytes, in complex with several associated proteins as well as the surrounding lipids, in its monomeric, dimeric and tetrameric forms without the use of cross-linking or chimeric mutations. These results point to the potential for studying isolated IMPs without the use of destabilizing detergents and thereby offer a pathway to analysis of wild-type and mutant membrane protein structure, function and pharmacodynamics

    The endoplasmic reticulum-associated Hsp40 DNAJB12 and Hsc70 cooperate to facilitate RMA1 E3-dependent degradation of nascent CFTR F508

    Get PDF
    A specialized Hsp40 protein, DNAJB12, was found to function on the cytoplasmic face of the ER with the RMA1 E3 ligase to regulate the folding efficiency of CFTR.Relative contributions of folding kinetics versus protein quality control (QC) activity in the partitioning of non-native proteins between life and death are not clear. Cystic fibrosis transmembrane conductance regulator (CFTR) biogenesis serves as an excellent model to study this question because folding of nascent CFTR is inefficient and deletion of F508 causes accumulation of CFTRΔF508 in a kinetically trapped, but foldable state. Herein, a novel endoplasmic reticulum (ER)-associated Hsp40, DNAJB12 (JB12) is demonstrated to play a role in control of CFTR folding efficiency. JB12 cooperates with cytosolic Hsc70 and the ubiquitin ligase RMA1 to target CFTR and CFTRΔF508 for degradation. Modest elevation of JB12 decreased nascent CFTR and CFTRΔF508 accumulation while increasing association of Hsc70 with ER forms of CFTR and the RMA1 E3 complex. Depletion of JB12 increased CFTR folding efficiency up to threefold and permitted a pool of CFTRΔF508 to fold and escape the ER. Introduction of the V510D misfolding suppressor mutation into CFTRΔF508 modestly increased folding efficiency, whereas combined inactivation of JB12 and suppression of intrinsic folding defects permitted CFTRΔF508 to fold at 50% of wild-type efficiency. Therapeutic correction of CFTRΔF508 misfolding in cystic fibrosis patients may require repair of defective folding kinetics and suppression of ER QC factors, such as JB12

    Deorphanization of Adhesion GPCRs overexpressed in Glioblastoma

    Get PDF
    G-protein-coupled receptors (GPCRs) represent, nowadays, one of the most productive source of drug targets. Some GPCRs members are, however, poorly studied and hence cannot be targeted for therapies yet. Among these, there are more than 90 GPCRs still orphan, i.e. with no described endogenous ligand and no clearly defined function. The orphan receptors CELSR2 and Frizzled4 (Fz4), have recently drawn the attention of the scientific community in virtue of their involvement in cancer progression. CELSR2 is an adhesion GPCR (class B GPCRs) overexpressed in glioblastoma. Until now, CELSR2 endogenous ligands have not yet been identified. Fz4 belongs to class F GPCRs and it is activated by the lipoproteins WNTs and Norrin. Misregulation of Fz4 activity is involved in tumor proliferation and cancer stem cell genesis in many types of malignancies, such as glioblastoma, colorectal and breast cancer. To date, the existence of low-molecular-weight organic molecules binding to and modulating Fz4 has not been reported. Thus, the identification of ligands/modulators of CELSR2 and Fz4 activity could pave the way for new therapeutic strategies to treat cancer. In this thesis, I attempted to identify CELSR2 and Fz4 ligands by using the “Pharmacological chaperone readout”, an innovative screening platform (PC-platform) that identifies ligands in virtue of their potency in affecting the tridimensional structure and the intracellular localization of a protein target. The first chapter of the thesis describes the results obtained in the attempt of identifying new ligands of Fz4 receptor. To achieve this aim, a cell line expressing a Fz4 mutant, Fz4-L501fsX533, was generated. This frameshift mutation is responsible in vivo for the occurrence of the Familial Exudative Vitreoretinopathy (FEVR), a pathology of the retina. The resulting mutant, here referred to as Fz4-FEVR, aggregates intracellularly in the Endoplasmic Reticulum (ER) without reaching the Plasma Membrane (PM) of the cell where, in contrast, the wt receptor localizes at steady state. To identify Fz4 wt modulators, a library of organic molecules has been screened for pharmacological chaperones of Fz4-FEVR, i.e. for molecules able to rescue the folding and correct localization of Fz4-FEVR at PM. Using such read-out, the organic molecule FzM1 has been thus identified as Fz4-FEVR pharmacological chaperone. The pharmacological chaperone FzM1 acts as allosteric inhibitor of the Fz4-wt receptor, binding directly to the wt receptor and inhibiting the signalling pathway Fz4 is involved in. I also performed a structure-activity relationship (SAR) analysis using FzM1 as lead to identify the first allosteric agonist of Fz4, FzM1.8. As discussed in the second chapter of this thesis, a specific PC-platform was developed for the identification of ligands of CELSR2. To achieve this aim, a misfolded version of CELSR2 was generated and then a library of metabolites was screened looking for compounds binding to the receptor and correcting its folding. Zebrafish eggs were used as biomass to obtain a library of metabolites which were screened for pharmacological chaperone efficiency. The outcome of the screening identified three compounds acting as modulators of CELSR2: cholesterol, PGE2 and ÎČ-carotene. They do not target the orthosteric binding site of CELSR2 but they are located in an hydrophobic region of the receptor at the interface between the TM-bundle and the lipid bilayer. The potential ability of these molecules to allosterically modulate CELSR2 function could have important implications both in physiological and pathological cell conditions

    SUMOylierung

    Get PDF
    Die vorliegende Arbeit untersucht die Frage, ob humanes CFTR-Protein (Cystic Fibrosis Transmembrane Conductance Regulator) sumoyliert wird und welche funktionelle Bedeutung die Sumoylierung des CFTRs hat. In der kaukasischen Bevölkerung gilt die Cystischen Fibrose (CF) oder Mukoviszidose als eine der hĂ€ufigsten Erbkrankheiten. Mutationen im CFTR-Gen, die zu einer drastischen BeeintrĂ€chtigung der Funktion des CFTR-Proteins fĂŒhren, bilden die molekulare Basis der Entstehung der CF. So sind hĂ€ufig die Reifung und der Transport von CFTR an die Plasmamembran betroffen. Diese Prozesse und verschiedene AktivitĂ€tszustĂ€nde im CFTR werden u.a. durch posttranslationale Modifikationen (z.B. Phosphorylierungen) spezifischer Konsensus-Motive reguliert. In diesem Zusammenhang fĂŒhrte eine genaue Betrachtung der PrimĂ€rsequenz von CFTR zur Identifikation von zwei hoch konservierten Motiven, die dem publizierten SUMO-Konsensus-Motiv (ψKxE, nachfolgend SUMO-Motiv genannt) an Position 447 und 1486, bezogen auf das zentrale Lysin, an welchem die Sumoylierung stattfindet, entsprachen. Die Sumoylierung, als eine Form der posttranslationale Modifikationen, wird durch kleine Ubiquitin-Ă€hnliche Proteine (small ubiquitin-like modifier, SUMO) vermittelt. Diese stellen eine Klasse von regulatorischen Proteinen dar, die die AktivitĂ€t des Zielproteins, dessen Lokalisation oder die Interaktion mit anderen Proteinen steuern. Damit ergab sich ein relevanter AnknĂŒpfungspunkt, die Rolle der bislang fĂŒr die CFTR-Regulation noch nicht beschriebenen Form der posttranslationalen Kontrolle durch Sumoylierung zu untersuchen. Als Resultat der hier beschriebenen Untersuchungen konnte die Sumoylierung als notwendiger Bestandteil der Regulation des intrazellulĂ€ren Transportes von CFTR identifiziert werden. Konkret wurde mit dem Yeast-Two-Hybrid-Verfahren gezeigt, dass CFTR-DomĂ€nen an den identifizierten SUMO-Motiven in der Nukleotid-bindedomĂ€ne 1 (NBD-1) und am C-Terminus von CFTR sumoyliert werden. Im SĂ€ugerzellsystem wurde die spezifische Sumoylierung kompletten CFTRs durch transiente Transfektion von CFTR- und SUMO-Expressionskonstrukten sowie in Zellen, welche CFTR und SUMO endogen exprimieren, verifiziert. Verhindert man weiterhin auf transiente Weise in SĂ€ugerzellen die Sumoylierung, durch Mutation der beiden SUMO-Motive oder durch Überexpression der SUMO-spezifischen Protease (SENP1), erreicht CFTR nicht die apikale Plasmamembran. Im Gegensatz zu F508, welches im ER verbleibt, wird CFTR, welches Mutationen in den SUMO-Motiven trĂ€gt, im Golgi-Apparat zurĂŒckgehalten. Diese Beobachtung konnte mit Hilfe der Dichtegradientenzentrifugation und anschließendem Nachweis der Proteine in den entsprechenden Fraktionen ĂŒber die Western-Blot-Methode gezeigt werden. Abschließend wurden am Beispiel eines zum SUMO-Motiv benachbart gelegenen Di-Leucin-Motives erste experimentelle Hinweise erhalten, wonach beide CFTR-Motive in der Interaktion mit dem fĂŒr den vesikulĂ€ren Transport funktionell wichtigem Adaptor-Protein (AP) Komplex zusammenwirken. Mit den vorliegenden Resultaten konnte erstmals die Hypothese formuliert werden, dass die hier beschriebene Sumoylierung eine Form der Modifikation ist, welche am Transport von CFTR an die Plasmamembran maßgeblich beteiligt ist
    corecore