35 results on '"Paavilainen VO"'
Search Results
2. Diastereomers of Coibamide A Show Altered Sec61 Client Selectivity and Ligand-Dependent Activity against Patient-Derived Glioma Stem-like Cells.
- Author
-
Mattos DR, Neves WD, Kitamura T, Pradhan R, Wan X, da Hora CC, Tranter D, Kazemi S, Yu X, Tripathy N, Paavilainen VO, McPhail KL, Oishi S, Badr CE, and Ishmael JE
- Abstract
Coibamide A (CbA) is a cyanobacterial lariat depsipeptide that selectively inhibits multiple secreted and integral membrane proteins from entering the endoplasmic reticulum secretory pathway through binding the alpha subunit of the Sec61 translocon. As a complex peptide-based macrocycle with 13 stereogenic centers, CbA is presumed to adopt a conformationally restricted orientation in the ligand-bound state, resulting in potent antitumor and antiangiogenic bioactivity. A stereochemical structure-activity relationship for CbA was previously defined based on cytotoxicity against established cancer cell lines. However, the ability of synthetic isomers to inhibit the biosynthesis of specific Sec61 substrates was unknown. Here, we report that two less toxic diastereomers of CbA, [L-Hiv
2 ]-CbA and [L-Hiv2 , L-MeAla11 ]-CbA, are pharmacologically active Sec61 inhibitors. Both compounds inhibited the expression of a secreted reporter ( Gaussia luciferase), VEGF-A, and a Type 1 membrane protein (VCAM1), while [L-Hiv2 ]-CbA also decreased the expression of ICAM1 and BiP/GRP78. Analysis of 43 different chemokines in the secretome of SF-268 glioblastoma cells revealed different inhibitory profiles for the two diastereomers. When the cytotoxic potential of CbA compounds was compared against a panel of patient-derived glioblastoma stem-like cells (GSCs), Sec61 inhibitors were remarkably toxic to five of the six GSCs tested. Each ligand showed a distinct cytotoxic potency and selectivity pattern for CbA-sensitive GSCs, with IC50 values ranging from subnanomolar to low micromolar concentrations. Together, these findings highlight the extreme sensitivity of GSCs to Sec61 modulation and the importance of ligand stereochemistry in determining the spectrum of inhibited Sec61 client proteins., Competing Interests: The authors declare no competing financial interest., (© 2024 The Authors. Published by American Chemical Society.)- Published
- 2024
- Full Text
- View/download PDF
3. The subset of peroxisomal tail-anchored proteins do not reach peroxisomes via ER, instead mitochondria can be involved.
- Author
-
Somborac T, Lutfullahoglu Bal G, Fatima K, Vihinen H, Paatero A, Jokitalo E, Paavilainen VO, and Konovalova S
- Subjects
- Animals, Endoplasmic Reticulum metabolism, Intracellular Membranes metabolism, Mitochondria metabolism, Mammals metabolism, Peroxisomes metabolism, Membrane Proteins metabolism
- Abstract
Peroxisomes are membrane-enclosed organelles with important roles in fatty acid breakdown, bile acid synthesis and biosynthesis of sterols and ether lipids. Defects in peroxisomes result in severe genetic diseases, such as Zellweger syndrome and neonatal adrenoleukodystrophy. However, many aspects of peroxisomal biogenesis are not well understood. Here we investigated delivery of tail-anchored (TA) proteins to peroxisomes in mammalian cells. Using glycosylation assays we showed that peroxisomal TA proteins do not enter the endoplasmic reticulum (ER) in both wild type (WT) and peroxisome-lacking cells. We observed that in cells lacking the essential peroxisome biogenesis factor, PEX19, peroxisomal TA proteins localize mainly to mitochondria. Finally, to investigate peroxisomal TA protein targeting in cells with fully functional peroxisomes we used a proximity biotinylation approach. We showed that while ER-targeted TA construct was exclusively inserted into the ER, peroxisome-targeted TA construct was inserted to both peroxisomes and mitochondria. Thus, in contrast to previous studies, our data suggest that some peroxisomal TA proteins do not insert to the ER prior to their delivery to peroxisomes, instead, mitochondria can be involved., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Somborac et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
4. Signal peptide mimicry primes Sec61 for client-selective inhibition.
- Author
-
Rehan S, Tranter D, Sharp PP, Craven GB, Lowe E, Anderl JL, Muchamuel T, Abrishami V, Kuivanen S, Wenzell NA, Jennings A, Kalyanaraman C, Strandin T, Javanainen M, Vapalahti O, Jacobson MP, McMinn D, Kirk CJ, Huiskonen JT, Taunton J, and Paavilainen VO
- Subjects
- Animals, Mice, Protein Transport, SEC Translocation Channels chemistry, SEC Translocation Channels genetics, SEC Translocation Channels metabolism, Protein Biosynthesis, Protein Sorting Signals, Membrane Proteins metabolism
- Abstract
Preventing the biogenesis of disease-relevant proteins is an attractive therapeutic strategy, but attempts to target essential protein biogenesis factors have been hampered by excessive toxicity. Here we describe KZR-8445, a cyclic depsipeptide that targets the Sec61 translocon and selectively disrupts secretory and membrane protein biogenesis in a signal peptide-dependent manner. KZR-8445 potently inhibits the secretion of pro-inflammatory cytokines in primary immune cells and is highly efficacious in a mouse model of rheumatoid arthritis. A cryogenic electron microscopy structure reveals that KZR-8445 occupies the fully opened Se61 lateral gate and blocks access to the lumenal plug domain. KZR-8445 binding stabilizes the lateral gate helices in a manner that traps select signal peptides in the Sec61 channel and prevents their movement into the lipid bilayer. Our results establish a framework for the structure-guided discovery of novel therapeutics that selectively modulate Sec61-mediated protein biogenesis., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
5. TSignal: a transformer model for signal peptide prediction.
- Author
-
Dumitrescu A, Jokinen E, Paatero A, Kellosalo J, Paavilainen VO, and Lähdesmäki H
- Subjects
- Protein Transport, Benchmarking, Language, Protein Sorting Signals, Amino Acids
- Abstract
Motivation: Signal peptides (SPs) are short amino acid segments present at the N-terminus of newly synthesized proteins that facilitate protein translocation into the lumen of the endoplasmic reticulum, after which they are cleaved off. Specific regions of SPs influence the efficiency of protein translocation, and small changes in their primary structure can abolish protein secretion altogether. The lack of conserved motifs across SPs, sensitivity to mutations, and variability in the length of the peptides make SP prediction a challenging task that has been extensively pursued over the years., Results: We introduce TSignal, a deep transformer-based neural network architecture that utilizes BERT language models and dot-product attention techniques. TSignal predicts the presence of SPs and the cleavage site between the SP and the translocated mature protein. We use common benchmark datasets and show competitive accuracy in terms of SP presence prediction and state-of-the-art accuracy in terms of cleavage site prediction for most of the SP types and organism groups. We further illustrate that our fully data-driven trained model identifies useful biological information on heterogeneous test sequences., Availability and Implementation: TSignal is available at: https://github.com/Dumitrescu-Alexandru/TSignal., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
6. Nonstop mRNAs generate a ground state of mitochondrial gene expression noise.
- Author
-
Ng KY, Lutfullahoglu Bal G, Richter U, Safronov O, Paulin L, Dunn CD, Paavilainen VO, Richer J, Newman WG, Taylor RW, and Battersby BJ
- Abstract
A stop codon within the mRNA facilitates coordinated termination of protein synthesis, releasing the nascent polypeptide from the ribosome. This essential step in gene expression is impeded with transcripts lacking a stop codon, generating nonstop ribosome complexes. Here, we use deep sequencing to investigate sources of nonstop mRNAs generated from the human mitochondrial genome. We identify diverse types of nonstop mRNAs on mitochondrial ribosomes that are resistant to translation termination by canonical release factors. Failure to resolve these aberrations by the mitochondrial release factor in rescue (MTRFR) imparts a negative regulatory effect on protein synthesis that is associated with human disease. Our findings reveal a source of underlying noise in mitochondrial gene expression and the importance of responsive ribosome quality control mechanisms for cell fitness and human health.
- Published
- 2022
- Full Text
- View/download PDF
7. Snapshots of actin and tubulin folding inside the TRiC chaperonin.
- Author
-
Kelly JJ, Tranter D, Pardon E, Chi G, Kramer H, Happonen L, Knee KM, Janz JM, Steyaert J, Bulawa C, Paavilainen VO, Huiskonen JT, and Yue WW
- Subjects
- Chaperonin Containing TCP-1 metabolism, Chaperonins chemistry, Chaperonins metabolism, Humans, Male, Peptides, Protein Folding, Actins metabolism, Tubulin metabolism
- Abstract
The integrity of a cell's proteome depends on correct folding of polypeptides by chaperonins. The chaperonin TCP-1 ring complex (TRiC) acts as obligate folder for >10% of cytosolic proteins, including he cytoskeletal proteins actin and tubulin. Although its architecture and how it recognizes folding substrates are emerging from structural studies, the subsequent fate of substrates inside the TRiC chamber is not defined. We trapped endogenous human TRiC with substrates (actin, tubulin) and cochaperone (PhLP2A) at different folding stages, for structure determination by cryo-EM. The already-folded regions of client proteins are anchored at the chamber wall, positioning unstructured regions toward the central space to achieve their native fold. Substrates engage with different sections of the chamber during the folding cycle, coupled to TRiC open-and-close transitions. Further, the cochaperone PhLP2A modulates folding, acting as a molecular strut between substrate and TRiC chamber. Our structural snapshots piece together an emerging model of client protein folding within TRiC., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
8. Carbonic anhydrase seven bundles filamentous actin and regulates dendritic spine morphology and density.
- Author
-
Bertling E, Blaesse P, Seja P, Kremneva E, Gateva G, Virtanen MA, Summanen M, Spoljaric I, Uvarov P, Blaesse M, Paavilainen VO, Vutskits L, Kaila K, Hotulainen P, and Ruusuvuori E
- Subjects
- Actin Cytoskeleton metabolism, Dendritic Spines metabolism, Hippocampus metabolism, Neurons metabolism, Actins genetics, Actins metabolism, Carbonic Anhydrases genetics
- Abstract
Intracellular pH is a potent modulator of neuronal functions. By catalyzing (de)hydration of CO
2 , intracellular carbonic anhydrase (CAi ) isoforms CA2 and CA7 contribute to neuronal pH buffering and dynamics. The presence of two highly active isoforms in neurons suggests that they may serve isozyme-specific functions unrelated to CO2 -(de)hydration. Here, we show that CA7, unlike CA2, binds to filamentous actin, and its overexpression induces formation of thick actin bundles and membrane protrusions in fibroblasts. In CA7-overexpressing neurons, CA7 is enriched in dendritic spines, which leads to aberrant spine morphology. We identified amino acids unique to CA7 that are required for direct actin interactions, promoting actin filament bundling and spine targeting. Disruption of CA7 expression in neocortical neurons leads to higher spine density due to increased proportion of small spines. Thus, our work demonstrates highly distinct subcellular expression patterns of CA7 and CA2, and a novel, structural role of CA7., (© 2021 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)- Published
- 2021
- Full Text
- View/download PDF
9. Targeting of HER/ErbB family proteins using broad spectrum Sec61 inhibitors coibamide A and apratoxin A.
- Author
-
Kazemi S, Kawaguchi S, Badr CE, Mattos DR, Ruiz-Saenz A, Serrill JD, Moasser MM, Dolan BP, Paavilainen VO, Oishi S, McPhail KL, and Ishmael JE
- Subjects
- Cell Survival drug effects, Cell Survival physiology, Dose-Response Relationship, Drug, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, HEK293 Cells, Humans, MCF-7 Cells, SEC Translocation Channels metabolism, Depsipeptides administration & dosage, Drug Delivery Systems methods, SEC Translocation Channels antagonists & inhibitors
- Abstract
Coibamide A is a potent cancer cell toxin and one of a select group of natural products that inhibit protein entry into the secretory pathway via a direct inhibition of the Sec61 protein translocon. Many Sec61 client proteins are clinically relevant drug targets once trafficked to their final destination in or outside the cell, however the use of Sec61 inhibitors to block early biosynthesis of specific proteins is at a pre-clinical stage. In the present study we evaluated the action of coibamide A against human epidermal growth factor receptor (HER, ErbB) proteins in representative breast and lung cancer cell types. HERs were selected for this study as they represent a family of Sec61 clients that is frequently dysregulated in human cancers, including coibamide-sensitive cell types. Although coibamide A inhibits biogenesis of a broad range of Sec61 substrate proteins in a presumed substrate-nonselective manner, endogenous HER3 (ErbB-3) and EGFR (ErbB-1) proteins were more sensitive to coibamide A, and the related Sec61 inhibitor apratoxin A, than HER2 (ErbB-2). Despite this rank order of sensitivity (HER3 > EGFR > HER2), Sec61-dependent inhibition by coibamide A was sufficient to decrease cell surface expression of HER2. We report that coibamide A- or apratoxin A-mediated block of HER3 entry into the secretory pathway is unlikely to be mediated by the HER3 signal peptide alone. HER3 (G11L/S15L), that is fully resistant to the highly substrate-selective cotransin analogue CT8, was more resistant than wild-type HER3 but only at low coibamide A (3 nM) concentrations; HER3 (G11L/S15L) expression was inhibited by higher concentrations of either natural product. Time- and concentration-dependent decreases in HER protein expression induced a commensurate reduction in AKT/MAPK signaling in breast and lung cancer cell types and loss in cell viability. Coibamide A potentiated the cytotoxic efficacy of small molecule kinase inhibitors lapatinib and erlotinib in breast and lung cancer cell types, respectively. These data indicate that natural product modulators of Sec61 function have value as chemical probes to interrogate HER/ErbB signaling in treatment-resistant human cancers., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
10. Coibamide A Targets Sec61 to Prevent Biogenesis of Secretory and Membrane Proteins.
- Author
-
Tranter D, Paatero AO, Kawaguchi S, Kazemi S, Serrill JD, Kellosalo J, Vogel WK, Richter U, Mattos DR, Wan X, Thornburg CC, Oishi S, McPhail KL, Ishmael JE, and Paavilainen VO
- Subjects
- Binding Sites, Cells, Cultured, Depsipeptides metabolism, Humans, Membrane Proteins biosynthesis, Photoaffinity Labels chemistry, SEC Translocation Channels metabolism, Depsipeptides pharmacology, Membrane Proteins antagonists & inhibitors, SEC Translocation Channels drug effects
- Abstract
Coibamide A (CbA) is a marine natural product with potent antiproliferative activity against human cancer cells and a unique selectivity profile. Despite promising antitumor activity, the mechanism of cytotoxicity and specific cellular target of CbA remain unknown. Here, we develop an optimized synthetic CbA photoaffinity probe (photo-CbA) and use it to demonstrate that CbA directly targets the Sec61α subunit of the Sec61 protein translocon. CbA binding to Sec61 results in broad substrate-nonselective inhibition of ER protein import and potent cytotoxicity against specific cancer cell lines. CbA targets a lumenal cavity of Sec61 that is partially shared with known Sec61 inhibitors, yet profiling against resistance conferring Sec61α mutations identified from human HCT116 cells suggests a distinct binding mode for CbA. Specifically, despite conferring strong resistance to all previously known Sec61 inhibitors, the Sec61α mutant R66I remains sensitive to CbA. A further unbiased screen for Sec61α resistance mutations identified the CbA-resistant mutation S71P, which confirms nonidentical binding sites for CbA and apratoxin A and supports the susceptibility of the Sec61 plug region for channel inhibition. Remarkably, CbA, apratoxin A, and ipomoeassin F do not display comparable patterns of potency and selectivity in the NCI60 panel of human cancer cell lines. Our work connecting CbA activity with selective prevention of secretory and membrane protein biogenesis by inhibition of Sec61 opens up possibilities for developing new Sec61 inhibitors with improved drug-like properties that are based on the coibamide pharmacophore.
- Published
- 2020
- Full Text
- View/download PDF
11. Protein translocation and retro-translocation across the endoplasmic reticulum are crucial to inflammatory effector CD4 + T cell function.
- Author
-
Pradeep Yeola A, Akbar I, Baillargeon J, Mercy Ignatius Arokia Doss P, Paavilainen VO, and Rangachari M
- Subjects
- Animals, Cell Differentiation immunology, Central Nervous System immunology, Cytokines immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Interferon-gamma immunology, Interleukin-17 immunology, Mice, Mice, Inbred C57BL, Multiple Sclerosis immunology, Th1 Cells immunology, Th17 Cells immunology, CD4-Positive T-Lymphocytes immunology, Endoplasmic Reticulum immunology, Inflammation immunology, Protein Transport immunology
- Abstract
Effector CD4
+ T cells can be classified by the cytokines they secrete, with T helper 1 (Th1) cells generating interferon (IFN)γ and Th17 cells secreting interleukin (IL)-17. Both Th1 and Th17 cells are strongly implicated in the initiation and chronicity of autoimmune diseases such as multiple sclerosis. The endoplasmic reticulum (ER) has been implicated as a potentially crucial site in regulating CD4+ T cell function. Secretory and transmembrane proteins are shuttled into the ER via the Sec61 translocon, where they undergo appropriate folding; misfolded proteins are retro-translocated from the ER in a p97-dependent manner. Here, we provide evidence that both processes are crucial to the secretion of inflammatory cytokines from effector CD4+ T cells. The pan-ER inhibitor eeeyarestatin-1 (ESI), which interferes with both Sec61 translocation and p97 retro-translocation, inhibited secretion of interferon (IFN)γ, interleukin (IL)-2 and tumor necrosis factor (TNF)α from Th1 cells in a dose-dependent manner. Selective inhibition of Sec61 by Apratoxin A (ApraA) revealed that ER translocation is crucial for Th1 cytokine secretion, while inhibition of p97 by NMS-873 also inhibited Th1 function, albeit to a lesser degree. By contrast, none of ESI, ApraA or NMS-873 could significantly reduce IL-17 secretion from Th17 cells. ApraA, but not NMS-873, reduced phosphorylation of Stat1 in Th1 cells, indicating the involvement of ER translocation in Th1 differentiation pathways. ApraA had modest effects on activation of the Th17 transcription factor Stat3, while NMS-873 had no effect. Interestingly, NMS-873 was able to reduce disease severity in CD4+ T cell-driven experimental autoimmune encephalomyelitis (EAE). Together, our data indicate that CD4+ T cell function, and Th1 cell function in particular, is dependent on protein translocation and dislocation across the ER., Competing Interests: Declaration of Competing Interest M.R. has performed educational activities for Biogen Canada and is the lead investigator on a research contract with Remedy Pharmaceuticals. These activities are unrelated to the work presented in this manuscript., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
12. Natural products as modulators of eukaryotic protein secretion.
- Author
-
Luesch H and Paavilainen VO
- Subjects
- Animals, Biological Products chemistry, Calcium metabolism, Endoplasmic Reticulum Stress drug effects, Endoplasmic Reticulum Stress physiology, Eukaryotic Cells metabolism, Humans, Protein Transport drug effects, RNA Splicing drug effects, RNA Splicing physiology, Biological Products pharmacology, Eukaryotic Cells drug effects, Proteins metabolism
- Abstract
Covering: up to the end of 2019Diverse natural product small molecules have allowed critical insights into processes that govern eukaryotic cells' ability to secrete cytosolically synthesized secretory proteins into their surroundings or to insert newly synthesized integral membrane proteins into the lipid bilayer of the endoplasmic reticulum. In addition, many components of the endoplasmic reticulum, required for protein homeostasis or other processes such as lipid metabolism or maintenance of calcium homeostasis, are being investigated for their potential in modulating human disease conditions such as cancer, neurodegenerative conditions and diabetes. In this review, we cover recent findings up to the end of 2019 on natural products that influence protein secretion or impact ER protein homeostasis, and serve as powerful chemical tools to understand protein flux through the mammalian secretory pathway and as leads for the discovery of new therapeutics.
- Published
- 2020
- Full Text
- View/download PDF
13. Kendomycin Cytotoxicity against Bacterial, Fungal, and Mammalian Cells Is Due to Cation Chelation.
- Author
-
Tranter D, Filipuzzi I, Lochmann T, Knapp B, Kellosalo J, Estoppey D, Pistorius D, Meissner A, Paavilainen VO, and Hoepfner D
- Subjects
- Cations, Cell Line, Copper metabolism, Iron metabolism, Leupeptins pharmacology, Microbial Sensitivity Tests, Mutagenesis, Rifabutin pharmacology, Yeasts drug effects, Anti-Bacterial Agents pharmacology, Antibiotics, Antineoplastic pharmacology, Antifungal Agents pharmacology, Bacteria drug effects, Chelating Agents pharmacology, Fungi drug effects, Rifabutin analogs & derivatives
- Abstract
Kendomycin is a small-molecule natural product that has gained significant attention due to reported cytotoxicity against pathogenic bacteria and fungi as well as a number of cancer cell lines. Despite significant biomedical interest and attempts to reveal its mechanism of action, the cellular target of kendomycin remains disputed. Herein it is shown that kendomycin induces cellular responses indicative of cation stress comparable to the effects of established iron chelators. Furthermore, addition of excess iron and copper attenuated kendomycin cytotoxicity in bacteria, yeast, and mammalian cells. Finally, NMR analysis demonstrated a direct interaction with cations, corroborating a close link between the observed kendomycin polypharmacology across different species and modulation of iron and/or copper levels.
- Published
- 2020
- Full Text
- View/download PDF
14. Current Progress on Equilibrative Nucleoside Transporter Function and Inhibitor Design.
- Author
-
Rehan S, Shahid S, Salminen TA, Jaakola VP, and Paavilainen VO
- Subjects
- Amino Acid Sequence, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Equilibrative Nucleoside Transport Proteins antagonists & inhibitors, Equilibrative Nucleoside Transport Proteins genetics, Humans, Ligands, Molecular Structure, Mutation, Protein Binding, Structure-Activity Relationship, Drug Design, Drug Discovery methods, Equilibrative Nucleoside Transport Proteins chemistry
- Abstract
Physiological nucleosides are used for the synthesis of DNA, RNA, and ATP in the cell and serve as universal mammalian signaling molecules that regulate physiological processes such as vasodilation and platelet aggregation by engaging with cell surface receptors. The same pathways that allow uptake of physiological nucleosides mediate the cellular import of synthetic nucleoside analogs used against cancer, HIV, and other viral diseases. Physiological nucleosides and nucleoside drugs are imported by two families of nucleoside transporters: the SLC28 concentrative nucleoside transporters (CNTs) and SLC29 equilibrative nucleoside transporters (ENTs). The four human ENT paralogs are expressed in distinct tissues, localize to different subcellular sites, and transport a variety of different molecules. Here we provide an overview of the known structure-function relationships of the ENT family with a focus on ligand binding and transport in the context of a new hENT1 homology model. We provide a generic residue numbering system for the different ENTs to facilitate the interpretation of mutational data produced using different ENT homologs. The discovery of paralog-selective small-molecule modulators is highly relevant for the design of new therapies and for uncovering the functions of poorly characterized ENT family members. Here, we discuss recent developments in the discovery of new paralog-selective small-molecule ENT inhibitors, including new natural product-inspired compounds. Recent progress in the ability to heterologously produce functional ENTs will allow us to gain insight into the structure and functions of different ENT family members as well as the rational discovery of highly selective inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
15. Wherever I may roam: organellar protein targeting and evolvability.
- Author
-
Dunn CD and Paavilainen VO
- Subjects
- Amino Acid Sequence genetics, Amoeba genetics, Amoeba metabolism, Endoplasmic Reticulum metabolism, Eukaryota metabolism, Evolution, Molecular, Mitochondria genetics, Molecular Chaperones genetics, Molecular Chaperones metabolism, Phylogeny, Protein Sorting Signals physiology, Protein Transport genetics, Protein Transport physiology, Eukaryota genetics, Gene Transfer, Horizontal genetics, Mitochondria metabolism, Protein Sorting Signals genetics
- Abstract
Many functions of eukaryotic cells are compartmentalized within membrane-bound organelles. One or more cis-encoded signals within a polypeptide sequence typically govern protein targeting to and within destination organelles. Perhaps unexpectedly, organelle targeting does not occur with high specificity, but instead is characterized by considerable degeneracy and inefficiency. Indeed, the same peptide signals can target proteins to more than one location, randomized sequences can easily direct proteins to organelles, and many enzymes appear to traverse different subcellular settings across eukaryotic phylogeny. We discuss the potential benefits provided by flexibility in organelle targeting, with a special emphasis on horizontally transferred and de novo proteins. Moreover, we consider how these new organelle residents can be protected and maintained before they contribute to the needs of the cell and promote fitness., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
16. Ipomoeassin F Binds Sec61α to Inhibit Protein Translocation.
- Author
-
Zong G, Hu Z, O'Keefe S, Tranter D, Iannotti MJ, Baron L, Hall B, Corfield K, Paatero AO, Henderson MJ, Roboti P, Zhou J, Sun X, Govindarajan M, Rohde JM, Blanchard N, Simmonds R, Inglese J, Du Y, Demangel C, High S, Paavilainen VO, and Shi WQ
- Subjects
- Binding Sites drug effects, Glycoconjugates chemistry, Humans, Molecular Structure, Protein Transport drug effects, SEC Translocation Channels metabolism, Glycoconjugates pharmacology, SEC Translocation Channels antagonists & inhibitors
- Abstract
Ipomoeassin F is a potent natural cytotoxin that inhibits growth of many tumor cell lines with single-digit nanomolar potency. However, its biological and pharmacological properties have remained largely unexplored. Building upon our earlier achievements in total synthesis and medicinal chemistry, we used chemical proteomics to identify Sec61α (protein transport protein Sec61 subunit alpha isoform 1), the pore-forming subunit of the Sec61 protein translocon, as a direct binding partner of ipomoeassin F in living cells. The interaction is specific and strong enough to survive lysis conditions, enabling a biotin analogue of ipomoeassin F to pull down Sec61α from live cells, yet it is also reversible, as judged by several experiments including fluorescent streptavidin staining, delayed competition in affinity pulldown, and inhibition of TNF biogenesis after washout. Sec61α forms the central subunit of the ER protein translocation complex, and the binding of ipomoeassin F results in a substantial, yet selective, inhibition of protein translocation in vitro and a broad ranging inhibition of protein secretion in live cells. Lastly, the unique resistance profile demonstrated by specific amino acid single-point mutations in Sec61α provides compelling evidence that Sec61α is the primary molecular target of ipomoeassin F and strongly suggests that the binding of this natural product to Sec61α is distinctive. Therefore, ipomoeassin F represents the first plant-derived, carbohydrate-based member of a novel structural class that offers new opportunities to explore Sec61α function and to further investigate its potential as a therapeutic target for drug discovery.
- Published
- 2019
- Full Text
- View/download PDF
17. Rapamycin-inspired macrocycles with new target specificity.
- Author
-
Guo Z, Hong SY, Wang J, Rehan S, Liu W, Peng H, Das M, Li W, Bhat S, Peiffer B, Ullman BR, Tse CM, Tarmakova Z, Schiene-Fischer C, Fischer G, Coe I, Paavilainen VO, Sun Z, and Liu JO
- Subjects
- Acute Kidney Injury metabolism, Acute Kidney Injury prevention & control, Animals, Cell Line, Human Umbilical Vein Endothelial Cells, Humans, Mice, Proteome metabolism, Reperfusion Injury metabolism, Reperfusion Injury prevention & control, Sirolimus chemistry, Sirolimus metabolism, Swine, TOR Serine-Threonine Kinases chemistry, TOR Serine-Threonine Kinases metabolism, Tacrolimus chemistry, Tacrolimus metabolism, Tacrolimus Binding Proteins chemistry, Tacrolimus Binding Proteins metabolism, Drug Discovery methods, Macrolides chemistry, Macrolides metabolism, Protective Agents chemistry, Protective Agents metabolism
- Abstract
Rapamycin and FK506 are macrocyclic natural products with an extraordinary mode of action, in which they form binary complexes with FK506-binding protein (FKBP) through a shared FKBP-binding domain before forming ternary complexes with their respective targets, mechanistic target of rapamycin (mTOR) and calcineurin, respectively. Inspired by this, we sought to build a rapamycin-like macromolecule library to target new cellular proteins by replacing the effector domain of rapamycin with a combinatorial library of oligopeptides. We developed a robust macrocyclization method using ring-closing metathesis and synthesized a 45,000-compound library of hybrid macrocycles (named rapafucins) using optimized FKBP-binding domains. Screening of the rapafucin library in human cells led to the discovery of rapadocin, an inhibitor of nucleoside uptake. Rapadocin is a potent, isoform-specific and FKBP-dependent inhibitor of the equilibrative nucleoside transporter 1 and is efficacious in an animal model of kidney ischaemia reperfusion injury. Together, these results demonstrate that rapafucins are a new class of chemical probes and drug leads that can expand the repertoire of protein targets well beyond mTOR and calcineurin.
- Published
- 2019
- Full Text
- View/download PDF
18. Proteomics Reveals Scope of Mycolactone-mediated Sec61 Blockade and Distinctive Stress Signature.
- Author
-
Morel JD, Paatero AO, Wei J, Yewdell JW, Guenin-Macé L, Van Haver D, Impens F, Pietrosemoli N, Paavilainen VO, and Demangel C
- Subjects
- Animals, Endoplasmic Reticulum Stress drug effects, Membrane Proteins metabolism, Mice, Substrate Specificity drug effects, Transcription Factors metabolism, Transcription, Genetic drug effects, Up-Regulation drug effects, Viral Proteins metabolism, Macrolides pharmacology, Proteomics methods, SEC Translocation Channels metabolism, Stress, Physiological drug effects
- Abstract
Mycolactone is a bacteria-derived macrolide that blocks the biogenesis of a large array of secretory and integral transmembrane proteins (TMP) through potent inhibition of the Sec61 translocon. Here, we used quantitative proteomics to delineate the direct and indirect effects of mycolactone-mediated Sec61 blockade in living cells. In T lymphocytes, dendritic cells and sensory neurons, Sec61 substrates downregulated by mycolactone were in order of incidence: secretory proteins (with a signal peptide but no transmembrane domain), TMPs with a signal peptide (Type I) and TMPs without signal peptide and a cytosolic N terminus (Type II). TMPs without a signal peptide and the opposite N terminus topology (Type III) were refractory to mycolactone inhibition. This rule applied comparably to single- and multi-pass TMPs, and extended to exogenous viral proteins. Parallel to its broad-spectrum inhibition of Sec61-mediated protein translocation, mycolactone rapidly induced cytosolic chaperones Hsp70/Hsp90. Moreover, it activated an atypical endoplasmic reticulum stress response, differing from conventional unfolded protein response by the down-regulation of Bip. In addition to refining our mechanistic understanding of Sec61 inhibition by mycolactone, our findings thus reveal that Sec61 blockade induces proteostatic stress in the cytosol and the endoplasmic reticulum., (© 2018 Morel et al.)
- Published
- 2018
- Full Text
- View/download PDF
19. ASD-Associated De Novo Mutations in Five Actin Regulators Show Both Shared and Distinct Defects in Dendritic Spines and Inhibitory Synapses in Cultured Hippocampal Neurons.
- Author
-
Hlushchenko I, Khanal P, Abouelezz A, Paavilainen VO, and Hotulainen P
- Abstract
Many actin cytoskeleton-regulating proteins control dendritic spine morphology and density, which are cellular features often altered in autism spectrum disorder (ASD). Recent studies using animal models show that autism-related behavior can be rescued by either manipulating actin regulators or by reversing dendritic spine density or morphology. Based on these studies, the actin cytoskeleton is a potential target pathway for developing new ASD treatments. Thus, it is important to understand how different ASD-associated actin regulators contribute to the regulation of dendritic spines and how ASD-associated mutations modulate this regulation. For this study, we selected five genes encoding different actin-regulating proteins and induced ASD-associated de novo missense mutations in these proteins. We assessed the functionality of the wild-type and mutated proteins by analyzing their subcellular localization, and by analyzing the dendritic spine phenotypes induced by the expression of these proteins. As the imbalance between excitation and inhibition has been suggested to have a central role in ASD, we additionally evaluated the density, size and subcellular localization of inhibitory synapses. Common for all the proteins studied was the enrichment in dendritic spines. ASD-associated mutations induced changes in the localization of α-actinin-4, which localized less to dendritic spines, and for SWAP-70 and SrGAP3, which localized more to dendritic spines. Among the wild-type proteins studied, only α-actinin-4 expression caused a significant change in dendritic spine morphology by increasing the mushroom spine density and decreasing thin spine density. We hypothesized that mutations associated with ASD shift dendritic spine morphology from mushroom to thin spines. An M554V mutation in α-actinin-4 ( ACTN4 ) resulted in the expected shift in dendritic spine morphology by increasing the density of thin spines. In addition, we observed a trend toward higher thin spine density with mutations in myosin IXb and SWAP-70. Myosin IIb and myosin IXb expression increased the proportion of inhibitory synapses in spines. The expression of mutated myosin IIb (Y265C), SrGAP3 (E469K), and SWAP-70 (L544F) induced variable changes in inhibitory synapses.
- Published
- 2018
- Full Text
- View/download PDF
20. Functional reconstitution of human equilibrative nucleoside transporter-1 into styrene maleic acid co-polymer lipid particles.
- Author
-
Rehan S, Paavilainen VO, and Jaakola VP
- Subjects
- Animals, Equilibrative Nucleoside Transporter 1 physiology, Humans, Membrane Lipids chemistry, Protein Stability, Sf9 Cells, Solubility, Equilibrative Nucleoside Transporter 1 chemistry, Maleates chemistry, Polystyrenes chemistry
- Abstract
The human equilibrative nucleoside transporter-1 (hENT1) is important for the entry of anti-cancer and anti-viral nucleoside analog therapeutics into the cell, and thus for their efficacy. Understanding of hENT1 structure-function relationship could assist with development of nucleoside analogs with better cellular uptake properties. However, structural and biophysical studies of hENT1 remain challenging as the hydrophobic nature of the protein leads to complete aggregation upon detergent-based membrane isolation. Here we report detergent-free reconstitution of the hENT1 transporter into styrene maleic acid co-polymer lipid particles (SMALPs) that form a native lipid disc. SMALP-purified hENT1, expressed in Sf9 insect cells binds a variety of ligands with a similar affinity as the protein in native membrane, and exhibits increased thermal stability compared to detergent-solubilized hENT1. hENT1-SMALPs purified using FLAG affinity M2 resin yielded ~0.4mg of active and homogenous protein per liter of culture as demonstrated by ligand binding, size-exclusion chromatography and SDS-PAGE analyses. Electrospray ionization mass spectrometry (ESI-MS) analysis showed that each hENT1 lipid disc contains 16 phosphatidylcholine (PC) and 2 phosphatidylethanolamine (PE) lipid molecules. Polyunsaturated lipids are specifically excluded from the hENT1 lipid discs, possibly reflecting a functional requirement for a dynamic lipid environment. Our work demonstrates that human nucleoside transporters can be extracted and purified without removal from their native lipid environment, opening up a wide range of possibilities for their biophysical and structural studies., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
21. Mycolactone subverts immunity by selectively blocking the Sec61 translocon.
- Author
-
Baron L, Paatero AO, Morel JD, Impens F, Guenin-Macé L, Saint-Auret S, Blanchard N, Dillmann R, Niang F, Pellegrini S, Taunton J, Paavilainen VO, and Demangel C
- Subjects
- Cell Adhesion drug effects, Cell Adhesion genetics, Cell Adhesion immunology, Humans, Jurkat Cells, Receptors, Interferon genetics, SEC Translocation Channels genetics, SEC Translocation Channels immunology, Signal Transduction genetics, Signal Transduction immunology, Interferon gamma Receptor, Macrolides pharmacology, Receptors, Interferon immunology, SEC Translocation Channels antagonists & inhibitors, Signal Transduction drug effects, T-Lymphocytes immunology
- Abstract
Mycolactone, an immunosuppressive macrolide released by the human pathogen Mycobacterium ulcerans, was previously shown to impair Sec61-dependent protein translocation, but the underlying molecular mechanism was not identified. In this study, we show that mycolactone directly targets the α subunit of the Sec61 translocon to block the production of secreted and integral membrane proteins with high potency. We identify a single-amino acid mutation conferring resistance to mycolactone, which localizes its interaction site near the lumenal plug of Sec61α. Quantitative proteomics reveals that during T cell activation, mycolactone-mediated Sec61 blockade affects a selective subset of secretory proteins including key signal-transmitting receptors and adhesion molecules. Expression of mutant Sec61α in mycolactone-treated T cells rescued their homing potential and effector functions. Furthermore, when expressed in macrophages, the mycolactone-resistant mutant restored IFN-γ receptor-mediated antimicrobial responses. Thus, our data provide definitive genetic evidence that Sec61 is the host receptor mediating the diverse immunomodulatory effects of mycolactone and identify Sec61 as a novel regulator of immune cell functions., (© 2016 Baron et al.)
- Published
- 2016
- Full Text
- View/download PDF
22. Apratoxin Kills Cells by Direct Blockade of the Sec61 Protein Translocation Channel.
- Author
-
Paatero AO, Kellosalo J, Dunyak BM, Almaliti J, Gestwicki JE, Gerwick WH, Taunton J, and Paavilainen VO
- Subjects
- Cell Death drug effects, Depsipeptides chemistry, Dose-Response Relationship, Drug, HCT116 Cells, Humans, Molecular Structure, Protein Transport drug effects, SEC Translocation Channels metabolism, Structure-Activity Relationship, Depsipeptides pharmacology, SEC Translocation Channels antagonists & inhibitors
- Abstract
Apratoxin A is a cytotoxic natural product that prevents the biogenesis of secretory and membrane proteins. Biochemically, apratoxin A inhibits cotranslational translocation into the ER, but its cellular target and mechanism of action have remained controversial. Here, we demonstrate that apratoxin A prevents protein translocation by directly targeting Sec61α, the central subunit of the protein translocation channel. Mutagenesis and competitive photo-crosslinking studies indicate that apratoxin A binds to the Sec61 lateral gate in a manner that differs from cotransin, a substrate-selective Sec61 inhibitor. In contrast to cotransin, apratoxin A does not exhibit a substrate-selective inhibitory mechanism, but blocks ER translocation of all tested Sec61 clients with similar potency. Our results suggest that multiple structurally unrelated natural products have evolved to target overlapping but non-identical binding sites on Sec61, thereby producing distinct biological outcomes., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
23. Prolonged and tunable residence time using reversible covalent kinase inhibitors.
- Author
-
Bradshaw JM, McFarland JM, Paavilainen VO, Bisconte A, Tam D, Phan VT, Romanov S, Finkle D, Shu J, Patel V, Ton T, Li X, Loughhead DG, Nunn PA, Karr DE, Gerritsen ME, Funk JO, Owens TD, Verner E, Brameld KA, Hill RJ, Goldstein DM, and Taunton J
- Subjects
- Acrylamides chemical synthesis, Agammaglobulinaemia Tyrosine Kinase, Animals, B-Lymphocytes enzymology, B-Lymphocytes pathology, Cell Line, Tumor, Crystallography, X-Ray, Cyanoacrylates chemical synthesis, Dasatinib, Female, Gene Expression, Humans, Ligands, Molecular Docking Simulation, Protein Kinase Inhibitors chemical synthesis, Protein Structure, Tertiary, Protein-Tyrosine Kinases chemistry, Protein-Tyrosine Kinases genetics, Pyrimidines pharmacokinetics, Rats, Rats, Sprague-Dawley, Recombinant Proteins chemistry, Recombinant Proteins genetics, Sf9 Cells, Spodoptera, Structure-Activity Relationship, Substrate Specificity, Thiazoles pharmacokinetics, Time Factors, Acrylamides pharmacokinetics, B-Lymphocytes drug effects, Cyanoacrylates pharmacokinetics, Protein Kinase Inhibitors pharmacokinetics, Protein-Tyrosine Kinases antagonists & inhibitors
- Abstract
Drugs with prolonged on-target residence times often show superior efficacy, yet general strategies for optimizing drug-target residence time are lacking. Here we made progress toward this elusive goal by targeting a noncatalytic cysteine in Bruton's tyrosine kinase (BTK) with reversible covalent inhibitors. Using an inverted orientation of the cysteine-reactive cyanoacrylamide electrophile, we identified potent and selective BTK inhibitors that demonstrated biochemical residence times spanning from minutes to 7 d. An inverted cyanoacrylamide with prolonged residence time in vivo remained bound to BTK for more than 18 h after clearance from the circulation. The inverted cyanoacrylamide strategy was further used to discover fibroblast growth factor receptor (FGFR) kinase inhibitors with residence times of several days, demonstrating the generalizability of the approach. Targeting of noncatalytic cysteines with inverted cyanoacrylamides may serve as a broadly applicable platform that facilitates 'residence time by design', the ability to modulate and improve the duration of target engagement in vivo.
- Published
- 2015
- Full Text
- View/download PDF
24. An allosteric Sec61 inhibitor traps nascent transmembrane helices at the lateral gate.
- Author
-
Mackinnon AL, Paavilainen VO, Sharma A, Hegde RS, and Taunton J
- Subjects
- Animals, Binding Sites, Cell Membrane metabolism, Cross-Linking Reagents pharmacology, Dose-Response Relationship, Drug, HCT116 Cells, HEK293 Cells, Humans, Membrane Proteins chemistry, Membrane Proteins genetics, Membrane Proteins metabolism, Molecular Docking Simulation, Mutation, Peptides, Cyclic metabolism, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Protein Transport, SEC Translocation Channels, Structure-Activity Relationship, Transfection, Tumor Necrosis Factor-alpha metabolism, Cell Membrane drug effects, Membrane Proteins antagonists & inhibitors, Peptides, Cyclic pharmacology
- Abstract
Membrane protein biogenesis requires the coordinated movement of hydrophobic transmembrane domains (TMD) from the cytosolic vestibule of the Sec61 channel into the lipid bilayer. Molecular insight into TMD integration has been hampered by the difficulty of characterizing intermediates during this intrinsically dynamic process. In this study, we show that cotransin, a substrate-selective Sec61 inhibitor, traps nascent TMDs in the cytosolic vestibule, permitting detailed interrogation of an early pre-integration intermediate. Site-specific crosslinking revealed the pre-integrated TMD docked to Sec61 near the cytosolic tip of the lateral gate. Escape from cotransin-arrest depends not only on cotransin concentration, but also on the biophysical properties of the TMD. Genetic selection of cotransin-resistant cancer cells uncovered multiple mutations clustered near the lumenal plug of Sec61α, thus revealing cotransin's likely site of action. Our results suggest that TMD/lateral gate interactions facilitate TMD transfer into the membrane, a process that is allosterically modulated by cotransin binding to the plug. DOI: http://dx.doi.org/10.7554/eLife.01483.001., Competing Interests: The authors declare that no competing interests exist.
- Published
- 2014
- Full Text
- View/download PDF
25. Electrophilic fragment-based design of reversible covalent kinase inhibitors.
- Author
-
Miller RM, Paavilainen VO, Krishnan S, Serafimova IM, and Taunton J
- Subjects
- Acrylamide chemical synthesis, Acrylamide chemistry, Cysteine chemistry, Dose-Response Relationship, Drug, Humans, Ligands, Models, Molecular, Molecular Structure, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Ribosomal Protein S6 Kinases, 90-kDa metabolism, Structure-Activity Relationship, Sulfhydryl Compounds chemistry, Acrylamide pharmacology, Protein Kinase Inhibitors pharmacology, Ribosomal Protein S6 Kinases, 90-kDa antagonists & inhibitors
- Abstract
Fragment-based ligand design and covalent targeting of noncatalytic cysteines have been employed to develop potent and selective kinase inhibitors. Here, we combine these approaches, starting with a panel of low-molecular-weight, heteroaryl-susbstituted cyanoacrylamides, which we have previously shown to form reversible covalent bonds with cysteine thiols. Using this strategy, we identify electrophilic fragments with sufficient ligand efficiency and selectivity to serve as starting points for the first reported inhibitors of the MSK1 C-terminal kinase domain. Guided by X-ray co-crystal structures, indazole fragment 1 was elaborated to afford 12 (RMM-46), a reversible covalent inhibitor that exhibits high ligand efficiency and selectivity for MSK/RSK-family kinases. At nanomolar concentrations, 12 blocked activation of cellular MSK and RSK, as well as downstream phosphorylation of the critical transcription factor, CREB.
- Published
- 2013
- Full Text
- View/download PDF
26. Proteostasis modulators with discriminating taste.
- Author
-
Paavilainen VO and Taunton J
- Abstract
Small molecules that perturb protein homeostasis are used as cancer therapeutics and as antibiotics to treat bacterial infections. In a recent issue of Cell, Kannan and colleagues describe an intriguing mechanism that enables ribosome-targeted macrolides to selectively remodel the bacterial proteome. This finding suggests the exciting possibility of targeting additional proteostasis regulators in a substrate-selective manner., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
27. Structure of the actin-depolymerizing factor homology domain in complex with actin.
- Author
-
Paavilainen VO, Oksanen E, Goldman A, and Lappalainen P
- Subjects
- Actin Cytoskeleton chemistry, Actin Depolymerizing Factors chemistry, Actins chemistry, Adenosine Triphosphate metabolism, Amino Acid Sequence, Animals, Conserved Sequence, Crystallography, X-Ray, Gelsolin chemistry, Mice, Models, Molecular, Molecular Sequence Data, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Actins metabolism, Destrin chemistry, Microfilament Proteins chemistry, Microfilament Proteins metabolism, Sequence Homology, Amino Acid
- Abstract
Actin dynamics provide the driving force for many cellular processes including motility and endocytosis. Among the central cytoskeletal regulators are actin-depolymerizing factor (ADF)/cofilin, which depolymerizes actin filaments, and twinfilin, which sequesters actin monomers and caps filament barbed ends. Both interact with actin through an ADF homology (ADF-H) domain, which is also found in several other actin-binding proteins. However, in the absence of an atomic structure for the ADF-H domain in complex with actin, the mechanism by which these proteins interact with actin has remained unknown. Here, we present the crystal structure of twinfilin's C-terminal ADF-H domain in complex with an actin monomer. This domain binds between actin subdomains 1 and 3 through an interface that is conserved among ADF-H domain proteins. Based on this structure, we suggest a mechanism by which ADF/cofilin and twinfilin inhibit nucleotide exchange of actin monomers and present a model for how ADF/cofilin induces filament depolymerization by weakening intrafilament interactions.
- Published
- 2008
- Full Text
- View/download PDF
28. Missing-in-metastasis and IRSp53 deform PI(4,5)P2-rich membranes by an inverse BAR domain-like mechanism.
- Author
-
Mattila PK, Pykäläinen A, Saarikangas J, Paavilainen VO, Vihinen H, Jokitalo E, and Lappalainen P
- Subjects
- Actins ultrastructure, Binding Sites physiology, Cell Differentiation physiology, Cell Line, Tumor, Cell Movement physiology, Cell Surface Extensions ultrastructure, Humans, Microtubules metabolism, Models, Molecular, Protein Binding physiology, Protein Structure, Tertiary physiology, Pseudopodia ultrastructure, Actins metabolism, Cell Surface Extensions metabolism, Microfilament Proteins metabolism, Neoplasm Proteins metabolism, Nerve Tissue Proteins metabolism, Phosphatidylinositol 4,5-Diphosphate metabolism, Pseudopodia metabolism
- Abstract
The actin cytoskeleton plays a fundamental role in various motile and morphogenetic processes involving membrane dynamics. We show that actin-binding proteins MIM (missing-in-metastasis) and IRSp53 directly bind PI(4,5)P(2)-rich membranes and deform them into tubular structures. This activity resides in the N-terminal IRSp53/MIM domain (IMD) of these proteins, which is structurally related to membrane-tubulating BAR (Bin/amphiphysin/Rvs) domains. We found that because of a difference in the geometry of the PI(4,5)P(2)-binding site, IMDs induce a membrane curvature opposite that of BAR domains and deform membranes by binding to the interior of the tubule. This explains why IMD proteins induce plasma membrane protrusions rather than invaginations. We also provide evidence that the membrane-deforming activity of IMDs, instead of the previously proposed F-actin-bundling or GTPase-binding activities, is critical for the induction of the filopodia/microspikes in cultured mammalian cells. Together, these data reveal that interplay between actin dynamics and a novel membrane-deformation activity promotes cell motility and morphogenesis.
- Published
- 2007
- Full Text
- View/download PDF
29. Structural basis and evolutionary origin of actin filament capping by twinfilin.
- Author
-
Paavilainen VO, Hellman M, Helfer E, Bovellan M, Annila A, Carlier MF, Permi P, and Lappalainen P
- Subjects
- Animals, Drosophila, Protein Structure, Tertiary, Actin Capping Proteins metabolism, Actins metabolism, Drosophila Proteins chemistry, Drosophila Proteins metabolism, Evolution, Molecular, Microfilament Proteins chemistry, Microfilament Proteins metabolism, Models, Molecular
- Abstract
Dynamic reorganization of the actin cytoskeleton is essential for motile and morphological processes in all eukaryotic cells. One highly conserved protein that regulates actin dynamics is twinfilin, which both sequesters actin monomers and caps actin filament barbed ends. Twinfilin is composed of two ADF/cofilin-like domains, Twf-N and Twf-C. Here, we reveal by systematic domain-swapping/inactivation analysis that the two functional ADF-H domains of twinfilin are required for barbed-end capping and that Twf-C plays a critical role in this process. However, these domains are not functionally equivalent. NMR-structure and mutagenesis analyses, together with biochemical and motility assays showed that Twf-C, in addition to its binding to G-actin, interacts with the sides of actin filaments like ADF/cofilins, whereas Twf-N binds only G-actin. Our results indicate that during filament barbed-end capping, Twf-N interacts with the terminal actin subunit, whereas Twf-C binds between two adjacent subunits at the side of the filament. Thus, the domain requirement for actin filament capping by twinfilin is remarkably similar to that of gelsolin family proteins, suggesting the existence of a general barbed-end capping mechanism. Furthermore, we demonstrate that a synthetic protein consisting of duplicated ADF/cofilin domains caps actin filament barbed ends, providing evidence that the barbed-end capping activity of twinfilin arose through a duplication of an ancient ADF/cofilin-like domain.
- Published
- 2007
- Full Text
- View/download PDF
30. NMR assignment of the C-terminal ADF-H domain of an actin monomer binding protein, twinfilin.
- Author
-
Hellman M, Paavilainen VO, Annila A, Lappalainen P, and Permi P
- Subjects
- Amino Acids analysis, Binding Sites, Carbon Isotopes, Nitrogen Isotopes, Nuclear Magnetic Resonance, Biomolecular, Peptide Fragments chemistry, Actins metabolism, Microfilament Proteins chemistry, Microfilament Proteins metabolism, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins metabolism
- Published
- 2006
- Full Text
- View/download PDF
31. Solution structure of coactosin reveals structural homology to ADF/cofilin family proteins.
- Author
-
Hellman M, Paavilainen VO, Naumanen P, Lappalainen P, Annila A, and Permi P
- Subjects
- Actin Depolymerizing Factors, Amino Acid Sequence, Amino Acids, Acidic, Amino Acids, Basic, Animals, Conserved Sequence, Destrin, Mice, Models, Molecular, Molecular Sequence Data, Protein Structure, Secondary, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Solutions, Microfilament Proteins chemistry
- Abstract
Coactosin is a small (MW approximately 15 kDa) evolutionarily conserved actin filament binding protein. It displays remote sequence homology to ADF/cofilin proteins and to the ADF-H domains of twinfilin and Abp1/drebrin. However, biochemical analyses have demonstrated that coactosin has a very different role in actin dynamics from the ones of ADF/cofilin, twinfilin or Abp1/drebrin. To elucidate the molecular mechanism of coactosin/actin interaction, we determined the three-dimensional structure of mouse coactosin by multidimensional NMR spectroscopy. We find that the coactosin structure is homologous to ADF/cofilin and to the ADF-H domains of twinfilin. Furthermore, the regions that have been shown to be important for actin filament interactions in ADF/cofilins are structurally conserved in coactosin suggesting that these two proteins interact with F-actin through a conserved interface. Our analysis also identifies key structural differences between these proteins that may account for the differences in biochemical activities and cellular roles of these proteins., (Copyright 2004 Federation of European Biochemical Societies)
- Published
- 2004
- Full Text
- View/download PDF
32. Biological role and structural mechanism of twinfilin-capping protein interaction.
- Author
-
Falck S, Paavilainen VO, Wear MA, Grossmann JG, Cooper JA, and Lappalainen P
- Subjects
- Actin Depolymerizing Factors, Actins metabolism, Animals, Binding Sites, Destrin, Mice, Microfilament Proteins genetics, Models, Molecular, Protein Binding, Protein Structure, Quaternary, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Microfilament Proteins chemistry, Microfilament Proteins metabolism, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Twinfilin and capping protein (CP) are highly conserved actin-binding proteins that regulate cytoskeletal dynamics in organisms from yeast to mammals. Twinfilin binds actin monomer, while CP binds the barbed end of the actin filament. Remarkably, twinfilin and CP also bind directly to each other, but the mechanism and role of this interaction in actin dynamics are not defined. Here, we found that the binding of twinfilin to CP does not affect the binding of either protein to actin. Furthermore, site-directed mutagenesis studies revealed that the CP-binding site resides in the conserved C-terminal tail region of twinfilin. The solution structure of the twinfilin-CP complex supports these conclusions. In vivo, twinfilin's binding to both CP and actin monomer was found to be necessary for twinfilin's role in actin assembly dynamics, based on genetic studies with mutants that have defined biochemical functions. Our results support a novel model for how sequential interactions between actin monomers, twinfilin, CP, and actin filaments promote cytoskeletal dynamics.
- Published
- 2004
- Full Text
- View/download PDF
33. Regulation of cytoskeletal dynamics by actin-monomer-binding proteins.
- Author
-
Paavilainen VO, Bertling E, Falck S, and Lappalainen P
- Subjects
- Animals, Models, Biological, Protein Structure, Tertiary, Actins physiology, Cytoskeleton physiology, Microfilament Proteins physiology
- Abstract
The actin cytoskeleton is a vital component of several key cellular and developmental processes in eukaryotes. Many proteins that interact with filamentous and/or monomeric actin regulate the structure and dynamics of the actin cytoskeleton. Actin-filament-binding proteins control the nucleation, assembly, disassembly and crosslinking of actin filaments, whereas actin-monomer-binding proteins regulate the size, localization and dynamics of the large pool of unpolymerized actin in cells. In this article, we focus on recent advances in understanding how the six evolutionarily conserved actin-monomer-binding proteins - profilin, ADF/cofilin, twinfilin, Srv2/CAP, WASP/WAVE and verprolin/WIP - interact with actin monomers and regulate their incorporation into filament ends. We also present a model of how, together, these ubiquitous actin-monomer-binding proteins contribute to cytoskeletal dynamics and actin-dependent cellular processes.
- Published
- 2004
- Full Text
- View/download PDF
34. Structural conservation between the actin monomer-binding sites of twinfilin and actin-depolymerizing factor (ADF)/cofilin.
- Author
-
Paavilainen VO, Merckel MC, Falck S, Ojala PJ, Pohl E, Wilmanns M, and Lappalainen P
- Subjects
- Actin Depolymerizing Factors, Amino Acid Sequence, Amino Acid Substitution, Animals, Binding Sites, Cloning, Molecular, Conserved Sequence, Crystallography, X-Ray, Escherichia coli genetics, Escherichia coli metabolism, Evolution, Molecular, Kinetics, Mice, Microfilament Proteins chemistry, Microfilament Proteins genetics, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Protein Structure, Secondary, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Sequence Alignment, Sequence Homology, Amino Acid, Actins chemistry, Actins metabolism, Microfilament Proteins metabolism, Saccharomyces cerevisiae Proteins
- Abstract
Twinfilin is an evolutionarily conserved actin monomer-binding protein that regulates cytoskeletal dynamics in organisms from yeast to mammals. It is composed of two actin-depolymerization factor homology (ADF-H) domains that show approximately 20% sequence identity to ADF/cofilin proteins. In contrast to ADF/cofilins, which bind both G-actin and F-actin and promote filament depolymerization, twinfilin interacts only with G-actin. To elucidate the molecular mechanisms of twinfilin-actin monomer interaction, we determined the crystal structure of the N-terminal ADF-H domain of twinfilin and mapped its actin-binding site by site-directed mutagenesis. This domain has similar overall structure to ADF/cofilins, and the regions important for actin monomer binding in ADF/cofilins are especially well conserved in twinfilin. Mutagenesis studies show that the N-terminal ADF-H domain of twinfilin and ADF/cofilins also interact with actin monomers through similar interfaces, although the binding surface is slightly extended in twinfilin. In contrast, the regions important for actin-filament interactions in ADF/cofilins are structurally different in twinfilin. This explains the differences in actin-interactions (monomer versus filament binding) between twinfilin and ADF/cofilins. Taken together, our data show that the ADF-H domain is a structurally conserved actin-binding motif and that relatively small structural differences at the actin interfaces of this domain are responsible for the functional variation between the different classes of ADF-H domain proteins.
- Published
- 2002
- Full Text
- View/download PDF
35. The two ADF-H domains of twinfilin play functionally distinct roles in interactions with actin monomers.
- Author
-
Ojala PJ, Paavilainen VO, Vartiainen MK, Tuma R, Weeds AG, and Lappalainen P
- Subjects
- Actin Depolymerizing Factors, Adenosine Diphosphate metabolism, Adenosine Triphosphate metabolism, Amino Acid Sequence, Animals, Binding Sites, Macromolecular Substances, Mice, Microfilament Proteins genetics, Models, Biological, Molecular Sequence Data, Protein Binding, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Sequence Alignment, Actins metabolism, Microfilament Proteins chemistry, Microfilament Proteins metabolism, Protein Conformation
- Abstract
Twinfilin is a ubiquitous and abundant actin monomer-binding protein that is composed of two ADF-H domains. To elucidate the role of twinfilin in actin dynamics, we examined the interactions of mouse twinfilin and its isolated ADF-H domains with G-actin. Wild-type twinfilin binds ADP-G-actin with higher affinity (K(D) = 0.05 microM) than ATP-G-actin (K(D) = 0.47 microM) under physiological ionic conditions and forms a relatively stable (k(off) = 1.8 s(-1)) complex with ADP-G-actin. Data from native PAGE and size exclusion chromatography coupled with light scattering suggest that twinfilin competes with ADF/cofilin for the high-affinity binding site on actin monomers, although at higher concentrations, twinfilin, cofilin, and actin may also form a ternary complex. By systematic deletion analysis, we show that the actin-binding activity is located entirely in the two ADF-H domains of twinfilin. Individually, these domains compete for the same binding site on actin, but the C-terminal ADF-H domain, which has >10-fold higher affinity for ADP-G-actin, is almost entirely responsible for the ability of twinfilin to increase the amount of monomeric actin in cosedimentation assays. Isolated ADF-H domains associate with ADP-G-actin with rapid second-order kinetics, whereas the association of wild-type twinfilin with G-actin exhibits kinetics consistent with a two-step binding process. These data suggest that the association with an actin monomer induces a first-order conformational change within the twinfilin molecule. On the basis of these results, we propose a kinetic model for the role of twinfilin in actin dynamics and its possible function in cells.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.