Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Nat Chem Biol ; 19(9): 1054-1062, 2023 09.
Article in English | MEDLINE | ID: mdl-37169961

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.


Subject(s)
Membrane Proteins , Protein Sorting Signals , Animals , Mice , Protein Transport , Membrane Proteins/metabolism , SEC Translocation Channels/chemistry , SEC Translocation Channels/genetics , SEC Translocation Channels/metabolism , Protein Biosynthesis
2.
EMBO Rep ; 24(12): e57910, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37983950

ABSTRACT

Protein translocation across the endoplasmic reticulum (ER) membrane is an essential step during protein entry into the secretory pathway. The conserved Sec61 protein-conducting channel facilitates polypeptide translocation and coordinates cotranslational polypeptide-processing events. In cells, the majority of Sec61 is stably associated with a heterotetrameric membrane protein complex, the translocon-associated protein complex (TRAP), yet the mechanism by which TRAP assists in polypeptide translocation remains unknown. Here, we present the structure of the core Sec61/TRAP complex bound to a mammalian ribosome by cryogenic electron microscopy (cryo-EM). Ribosome interactions anchor the Sec61/TRAP complex in a conformation that renders the ER membrane locally thinner by significantly curving its lumenal leaflet. We propose that TRAP stabilizes the ribosome exit tunnel to assist nascent polypeptide insertion through Sec61 and provides a ratcheting mechanism into the ER lumen mediated by direct polypeptide interactions.


Subject(s)
Endoplasmic Reticulum , Membrane Proteins , Animals , SEC Translocation Channels/genetics , SEC Translocation Channels/metabolism , Membrane Proteins/genetics , Membrane Proteins/chemistry , Endoplasmic Reticulum/metabolism , Mammals/metabolism , Peptides/metabolism , Protein Transport
3.
J Nat Prod ; 83(4): 965-971, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32182062

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antifungal Agents/pharmacology , Bacteria/drug effects , Chelating Agents/pharmacology , Fungi/drug effects , Rifabutin/analogs & derivatives , Cations , Cell Line , Copper/metabolism , Iron/metabolism , Leupeptins/pharmacology , Microbial Sensitivity Tests , Mutagenesis , Rifabutin/pharmacology , Yeasts/drug effects
4.
J Am Chem Soc ; 141(21): 8450-8461, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31059257

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.


Subject(s)
Glycoconjugates/pharmacology , SEC Translocation Channels/antagonists & inhibitors , Binding Sites/drug effects , Glycoconjugates/chemistry , Humans , Molecular Structure , Protein Transport/drug effects , SEC Translocation Channels/metabolism
5.
Biophys J ; 112(11): 2315-2326, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28591604

ABSTRACT

G protein-coupled receptors (GPCRs) have evolved a seven-transmembrane helix framework that is responsive to a wide range of extracellular signals. An analysis of the interior packing of family A GPCR crystal structures reveals two clusters of highly packed residues that facilitate tight transmembrane helix association. These clusters are centered on amino acid positions 2.47 and 4.53, which are highly conserved as alanine and serine, respectively. Ala2.47 mediates the interaction between helices H1 and H2, while Ser4.53 mediates the interaction between helices H3 and H4. The helical interfaces outside of these clusters are lined with residues that are more loosely packed, a structural feature that facilitates motion of helices H5, H6, and H7, which is required for receptor activation. Mutation of the conserved small side chain at position 4.53 within packing cluster 2 is shown to disrupt the structure of the visual receptor rhodopsin, whereas sites in packing cluster 1 (e.g., positions 1.46 and 2.47) are more tolerant to mutation but affect the overall stability of the protein. These findings reveal a common structural scaffold of GPCRs that is important for receptor folding and activation.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , COS Cells , Hydrogen Bonding , Models, Molecular , Motion , Mutation , Protein Conformation , Protein Folding , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Rhodopsin/chemistry , Rhodopsin/genetics , Rhodopsin/metabolism
6.
ACS Pharmacol Transl Sci ; 7(6): 1823-1838, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38898945

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-Hiv2]-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.

7.
Nat Struct Mol Biol ; 29(5): 420-429, 2022 05.
Article in English | MEDLINE | ID: mdl-35449234

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.


Subject(s)
Actins , Tubulin , Actins/metabolism , Chaperonin Containing TCP-1/metabolism , Chaperonins/chemistry , Chaperonins/metabolism , Humans , Male , Peptides , Protein Folding , Tubulin/metabolism
8.
ACS Chem Biol ; 15(8): 2125-2136, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32608972

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.


Subject(s)
Depsipeptides/pharmacology , Membrane Proteins/antagonists & inhibitors , SEC Translocation Channels/drug effects , Binding Sites , Cells, Cultured , Depsipeptides/metabolism , Humans , Membrane Proteins/biosynthesis , Photoaffinity Labels/chemistry , SEC Translocation Channels/metabolism
9.
Methods Enzymol ; 556: 283-305, 2015.
Article in English | MEDLINE | ID: mdl-25857787

ABSTRACT

The large-scale expression of many membrane proteins, including the members of the G protein-coupled receptor superfamily, in a correctly folded and fully functional form remains a formidable challenge. In this chapter, we focus on the construction of stable mammalian cell lines to overcome this hurdle. First, we will outline the steps for establishing a tightly regulated gene expression system in human HEK293S cells. This system utilizes separate plasmids containing components of well-defined genetic control elements from the Escherichia coli tetracycline operon to control the powerful cytomegalovirus immediate early enhancer/promoter. Next, we describe the assembly of this expression system into HEK293S cells and a derivative cell line devoid of complex N-glycosylation. Finally, we describe methods for the growth of these cells lines in scalable suspension culture for the preparation of milligram amounts of recombinant protein.


Subject(s)
Cloning, Molecular/methods , Receptors, G-Protein-Coupled/genetics , Animals , Cell Culture Techniques/methods , Escherichia coli/genetics , Gene Expression Regulation , Glycosylation , HEK293 Cells/cytology , HEK293 Cells/metabolism , Humans , Operon , Receptors, G-Protein-Coupled/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Tetracycline/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL