Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Nat Chem ; 16(6): 913-921, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38531969

ABSTRACT

Post-translational modifications (PTMs) dynamically regulate cellular processes. Lysine undergoes a range of acylations, including malonylation, succinylation (SucK) and glutarylation (GluK). These PTMs increase the size of the lysine side chain and reverse its charge from +1 to -1 under physiological conditions, probably impacting protein structure and function. To understand the functional roles of these PTMs, homogeneously modified proteins are required for biochemical studies. While the site-specific encoding of PTMs and their mimics via genetic code expansion has facilitated the characterization of the functional roles of many PTMs, negatively charged lysine acylations have defied this approach. Here we describe site-specific incorporation of SucK and GluK into proteins via temporarily masking their negative charge through thioester derivatives. We prepare succinylated and glutarylated bacterial and mammalian target proteins, including non-refoldable multidomain proteins. This allows us to study how succinylation and glutarylation impact enzymatic activity of metabolic enzymes and regulate protein-DNA and protein-protein interactions in biological processes from replication to ubiquitin signalling.


Subject(s)
Genetic Code , Protein Processing, Post-Translational , Succinic Acid , Succinic Acid/metabolism , Succinic Acid/chemistry , Humans , Lysine/chemistry , Lysine/metabolism , Proteins/chemistry , Proteins/metabolism , Proteins/genetics , Glutarates/metabolism , Glutarates/chemistry
2.
Chem Sci ; 15(6): 2229-2235, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38332832

ABSTRACT

Dibenzoannulated cyclooctynes have emerged as valuable compounds for bioorthogonal reactions. They are commonly used in combination with azides in strain-promoted 1,3-dipolar cycloadditions. They are typically, however, unreactive towards 3,6-disubstituted tetrazines in inverse electron-demand Diels-Alder cycloadditions. Recently a dibenzoannulated bicyclo[6.1.0]nonyne derivative (DMBO) with a cyclopropane fused to the cyclooctyne core was described, which showed surprising reactivity towards tetrazines. To elucidate the unusual reactivity of DMBO, we performed density functional theory calculations and revealed that a tub-like structure in the transition state results in a much lower activation barrier than in the absence of cyclopropane fusion. The same transition state geometry is found for different cycloalkanes fused to the cyclooctyne core albeit higher activation barriers are observed for increased ring sizes. This conformation is energetically unfavored for previously known dibenzoannulated cyclooctynes and allows tetrazines and azides to approach DMBO from the face rather than the edge, a trajectory that was hitherto not observed for this class of activated dieno- and dipolarophiles.

3.
J Am Chem Soc ; 145(36): 19513-19517, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37642301

ABSTRACT

Azomethine imines are valuable substrates for chemical synthesis in organic solvents that often require anhydrous conditions. Here, we introduce C,N-cyclic-N'-acyl azomethine imines (AMIs) to bioorthogonal reactions in an aqueous environment. These AMIs are stable under physiological conditions and react rapidly (k2 = 0.1-250 M-1 s-1, depending on pH) and chemoselectively with isonitriles in the presence of biological nucleophiles, including thiols. Live-cell imaging of cell-surface-bound isonitriles underlines the biocompatibility of the AMI-isonitrile ligation, and simultaneous one-pot triple-protein labeling demonstrates its orthogonality to commonly used bioorthogonal reactions, such as the SPAAC and iEDDA ligations.


Subject(s)
Azo Compounds , Imines , Cell Membrane , Biology
4.
J Biol Chem ; 298(12): 102677, 2022 12.
Article in English | MEDLINE | ID: mdl-36336075

ABSTRACT

Cytokines of the interleukin 12 (IL-12) family are assembled combinatorially from shared α and ß subunits. A common theme is that human IL-12 family α subunits remain incompletely structured in isolation until they pair with a designate ß subunit. Accordingly, chaperones need to support and control specific assembly processes. It remains incompletely understood, which chaperones are involved in IL-12 family biogenesis. Here, we site-specifically introduce photocrosslinking amino acids into the IL-12 and IL-23 α subunits (IL-12α and IL-23α) for stabilization of transient chaperone-client complexes for mass spectrometry. Our analysis reveals that a large set of endoplasmic reticulum chaperones interacts with IL-12α and IL-23α. Among these chaperones, we focus on protein disulfide isomerase (PDI) family members and reveal IL-12 family subunits to be clients of several incompletely characterized PDIs. We find that different PDIs show selectivity for different cysteines in IL-12α and IL-23α. Despite this, PDI binding generally stabilizes unassembled IL-12α and IL-23α against degradation. In contrast, α:ß assembly appears robust, and only multiple simultaneous PDI depletions reduce IL-12 secretion. Our comprehensive analysis of the IL-12/IL-23 chaperone machinery reveals a hitherto uncharacterized role for several PDIs in this process. This extends our understanding of how cells accomplish the task of specific protein assembly reactions for signaling processes. Furthermore, our findings show that cytokine secretion can be modulated by targeting specific endoplasmic reticulum chaperones.


Subject(s)
Cytokines , Protein Disulfide-Isomerases , Humans , Interleukin-12 , Interleukin-23 , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Folding , Endoplasmic Reticulum
5.
J Am Chem Soc ; 144(29): 13118-13126, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35850488

ABSTRACT

Asparaginyl endopeptidases (AEPs) have recently been widely utilized for peptide and protein modification. Labeling is however restricted to protein termini, severely limiting flexibility and scope in creating diverse conjugates as needed for therapeutic and diagnostic applications. Here, we use genetic code expansion to site-specifically modify target proteins with an isopeptide-linked glycylglycine moiety that serves as an acceptor nucleophile in AEP-mediated transpeptidation with various probes containing a tripeptidic recognition motif. Our approach allows simple and flexible labeling of recombinant proteins at any internal site and leaves a minimal, entirely peptidic footprint (NGG) in the conjugation product. We show site-specific labeling of diverse target proteins with various biophysical probes, including dual labeling at an internal site and the N-terminus. Furthermore, we harness AEP-mediated transpeptidation for generation of ubiquitin- and ubiquitin-like-modifier conjugates bearing a native isopeptide bond and only one point mutation in the linker region.


Subject(s)
Cysteine Endopeptidases , Peptides , Cysteine Endopeptidases/metabolism , Recombinant Proteins/metabolism , Ubiquitin/genetics
6.
Angew Chem Int Ed Engl ; 61(10): e202111085, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34847623

ABSTRACT

Approaches for profiling protease substrates are critical for defining protease functions, but remain challenging tasks. We combine genetic code expansion, photocrosslinking and proteomics to identify substrates of the mitochondrial (mt) human caseinolytic protease P (hClpP). Site-specific incorporation of the diazirine-bearing amino acid DiazK into the inner proteolytic chamber of hClpP, followed by UV-irradiation of cells, allows to covalently trap substrate proteins of hClpP and to substantiate hClpP's major involvement in maintaining overall mt homeostasis. In addition to confirming many of the previously annotated hClpP substrates, our approach adds a diverse set of new proteins to the hClpP interactome. Importantly, our workflow allows identifying substrate dynamics upon application of external cues in an unbiased manner. Identification of unique hClpP-substrate proteins upon induction of mt oxidative stress, suggests that hClpP counteracts oxidative stress by processing of proteins that are involved in respiratory chain complex synthesis and maturation as well as in catabolic pathways.


Subject(s)
Cross-Linking Reagents/metabolism , Endopeptidase Clp/metabolism , Mitochondria/enzymology , Cross-Linking Reagents/chemistry , Endopeptidase Clp/chemistry , Humans , Models, Molecular , Molecular Structure , Photochemical Processes , Substrate Specificity
7.
Nat Commun ; 12(1): 6515, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764289

ABSTRACT

The post-translational modification of proteins with ubiquitin (Ub) and Ub-like modifiers (Ubls) represents one of the most important regulators in eukaryotic biology. Polymeric Ub/Ubl chains of distinct topologies control the activity, stability, interaction and localization of almost all cellular proteins and elicit a variety of biological outputs. Our ability to characterize the roles of distinct Ub/Ubl topologies and to identify enzymes and receptors that create, recognize and remove these modifications is however hampered by the difficulty to prepare them. Here we introduce a modular toolbox (Ubl-tools) that allows the stepwise assembly of Ub/Ubl chains in a flexible and user-defined manner facilitated by orthogonal sortase enzymes. We demonstrate the universality and applicability of Ubl-tools by generating distinctly linked Ub/Ubl hybrid chains, and investigate their role in DNA damage repair. Importantly, Ubl-tools guarantees straightforward access to target proteins, site-specifically modified with distinct homo- and heterotypic (including branched) Ub chains, providing a powerful approach for studying the functional impact of these complex modifications on cellular processes.


Subject(s)
Polymers/chemistry , Ubiquitin/metabolism , DNA Damage/genetics , DNA Damage/physiology , Humans , Protein Binding , Protein Processing, Post-Translational , Ubiquitin/genetics , Ubiquitination/genetics , Ubiquitination/physiology
8.
Article in English | MEDLINE | ID: mdl-34585143

ABSTRACT

Bioorthogonal chemistry represents a class of high-yielding chemical reactions that proceed rapidly and selectively in biological environments without side reactions towards endogenous functional groups. Rooted in the principles of physical organic chemistry, bioorthogonal reactions are intrinsically selective transformations not commonly found in biology. Key reactions include native chemical ligation and the Staudinger ligation, copper-catalysed azide-alkyne cycloaddition, strain-promoted [3 + 2] reactions, tetrazine ligation, metal-catalysed coupling reactions, oxime and hydrazone ligations as well as photoinducible bioorthogonal reactions. Bioorthogonal chemistry has significant overlap with the broader field of 'click chemistry' - high-yielding reactions that are wide in scope and simple to perform, as recently exemplified by sulfuryl fluoride exchange chemistry. The underlying mechanisms of these transformations and their optimal conditions are described in this Primer, followed by discussion of how bioorthogonal chemistry has become essential to the fields of biomedical imaging, medicinal chemistry, protein synthesis, polymer science, materials science and surface science. The applications of bioorthogonal chemistry are diverse and include genetic code expansion and metabolic engineering, drug target identification, antibody-drug conjugation and drug delivery. This Primer describes standards for reproducibility and data deposition, outlines how current limitations are driving new research directions and discusses new opportunities for applying bioorthogonal chemistry to emerging problems in biology and biomedicine.

9.
Nucleic Acids Res ; 49(11): e62, 2021 06 21.
Article in English | MEDLINE | ID: mdl-33684219

ABSTRACT

The genetic code of mammalian cells can be expanded to allow the incorporation of non-canonical amino acids (ncAAs) by suppressing in-frame amber stop codons (UAG) with an orthogonal pyrrolysyl-tRNA synthetase (PylRS)/tRNAPylCUA (PylT) pair. However, the feasibility of this approach is substantially hampered by unpredictable variations in incorporation efficiencies at different stop codon positions within target proteins. Here, we apply a proteomics-based approach to quantify ncAA incorporation rates at hundreds of endogenous amber stop codons in mammalian cells. With these data, we compute iPASS (Identification of Permissive Amber Sites for Suppression; available at www.bultmannlab.eu/tools/iPASS), a linear regression model to predict relative ncAA incorporation efficiencies depending on the surrounding sequence context. To verify iPASS, we develop a dual-fluorescence reporter for high-throughput flow-cytometry analysis that reproducibly yields context-specific ncAA incorporation efficiencies. We show that nucleotides up- and downstream of UAG synergistically influence ncAA incorporation efficiency independent of cell line and ncAA identity. Additionally, we demonstrate iPASS-guided optimization of ncAA incorporation rates by synonymous exchange of codons flanking the amber stop codon. This combination of in silico analysis followed by validation in living mammalian cells substantially simplifies identification as well as adaptation of sites within a target protein to confer high ncAA incorporation rates.


Subject(s)
Amino Acids/metabolism , Genetic Code , Animals , Cell Line , Codon , Codon, Terminator , Computer Simulation , Embryonic Stem Cells/metabolism , Flow Cytometry , Genes, Reporter , HEK293 Cells , Humans , Linear Models , Mice , Mutation , Proteomics
10.
Nat Commun ; 12(1): 1564, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692360

ABSTRACT

The lipid phosphatidylinositol-3-phosphate (PI3P) is a regulator of two fundamental but distinct cellular processes, endocytosis and autophagy, so its generation needs to be under precise temporal and spatial control. PI3P is generated by two complexes that both contain the lipid kinase VPS34: complex II on endosomes (VPS34/VPS15/Beclin 1/UVRAG), and complex I on autophagosomes (VPS34/VPS15/Beclin 1/ATG14L). The endosomal GTPase Rab5 binds complex II, but the mechanism of VPS34 activation by Rab5 has remained elusive, and no GTPase is known to bind complex I. Here we show that Rab5a-GTP recruits endocytic complex II to membranes and activates it by binding between the VPS34 C2 and VPS15 WD40 domains. Electron cryotomography of complex II on Rab5a-decorated vesicles shows that the VPS34 kinase domain is released from inhibition by VPS15 and hovers over the lipid bilayer, poised for catalysis. We also show that the GTPase Rab1a, which is known to be involved in autophagy, recruits and activates the autophagy-specific complex I, but not complex II. Both Rabs bind to the same VPS34 interface but in a manner unique for each. These findings reveal how VPS34 complexes are activated on membranes by specific Rab GTPases and how they are recruited to unique cellular locations.


Subject(s)
Cell Membrane/metabolism , Class III Phosphatidylinositol 3-Kinases/chemistry , Class III Phosphatidylinositol 3-Kinases/metabolism , rab1 GTP-Binding Proteins/chemistry , rab1 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/chemistry , rab5 GTP-Binding Proteins/metabolism , Beclin-1/chemistry , Beclin-1/genetics , Beclin-1/metabolism , Class III Phosphatidylinositol 3-Kinases/genetics , Endosomes/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Structure, Secondary , Tomography , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vacuolar Sorting Protein VPS15/chemistry , Vacuolar Sorting Protein VPS15/genetics , Vacuolar Sorting Protein VPS15/metabolism , rab1 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics
11.
Nat Commun ; 12(1): 460, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33469029

ABSTRACT

Legionella pneumophila infects eukaryotic cells by forming a replicative organelle - the Legionella containing vacuole. During this process, the bacterial protein DrrA/SidM is secreted and manipulates the activity and post-translational modification (PTM) states of the vesicular trafficking regulator Rab1. As a result, Rab1 is modified with an adenosine monophosphate (AMP), and this process is referred to as AMPylation. Here, we use a chemical approach to stabilise low-affinity Rab:DrrA complexes in a site-specific manner to gain insight into the molecular basis of the interaction between the Rab protein and the AMPylation domain of DrrA. The crystal structure of the Rab:DrrA complex reveals a previously unknown non-conventional Rab-binding site (NC-RBS). Biochemical characterisation demonstrates allosteric stimulation of the AMPylation activity of DrrA via Rab binding to the NC-RBS. We speculate that allosteric control of DrrA could in principle prevent random and potentially cytotoxic AMPylation in the host, thereby perhaps ensuring efficient infection by Legionella.


Subject(s)
Adenosine Monophosphate/metabolism , Bacterial Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Legionella pneumophila/pathogenicity , Legionnaires' Disease/pathology , rab1 GTP-Binding Proteins/metabolism , Allosteric Regulation , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/ultrastructure , Binding Sites/genetics , Crystallography, X-Ray , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/isolation & purification , Guanine Nucleotide Exchange Factors/ultrastructure , Guanosine Triphosphate/metabolism , Humans , Legionella pneumophila/metabolism , Legionnaires' Disease/microbiology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Phagocytosis , Protein Binding , Protein Processing, Post-Translational , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , rab1 GTP-Binding Proteins/genetics , rab1 GTP-Binding Proteins/isolation & purification , rab1 GTP-Binding Proteins/ultrastructure
12.
Nat Chem ; 12(11): 980-982, 2020 11.
Article in English | MEDLINE | ID: mdl-33077926
13.
Curr Opin Chem Biol ; 58: 112-120, 2020 10.
Article in English | MEDLINE | ID: mdl-32911429

ABSTRACT

In recent years it has become possible to genetically encode an expanded set of designer amino acids with tailored chemical and physical properties (dubbed unnatural amino acids, UAAs) into proteins in living cells by expanding the genetic code. Together with developments in chemistries that are amenable to and selective within physiological settings, these strategies have started to have a big impact on biological studies, as they enable exciting in cellulo applications. Here we highlight recent advances to covalently stabilize transient protein-protein interactions and capture enzyme substrate-complexes in living cells using proximity-triggered and residue-selective photo-induced crosslinking approaches. Furthermore, we describe recent efforts in controlling enzyme activity with photocaged UAAs and in extending their application to a variety of enzymatic scaffolds. In addition, we discuss the site-specific incorporation of UAAs mimicking post-translational modifications (PTMs) and approaches to generate natively-linked ubiquitin-protein conjugates to probe the role of PTMs in modulating complex cellular networks.


Subject(s)
Genetic Code , Protein Engineering/methods , Proteins/chemistry , Proteins/genetics , Cell Survival , Humans , Proteins/metabolism
15.
Nat Commun ; 11(1): 3727, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32694502

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

16.
Nat Commun ; 11(1): 1219, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139682

ABSTRACT

Methylation of a conserved lysine in C-terminal domain of the molecular chaperone Hsp90 was shown previously to affect its in vivo function. However, the underlying mechanism remained elusive. Through a combined experimental and computational approach, this study shows that this site is very sensitive to sidechain modifications and crucial for Hsp90 activity in vitro and in vivo. Our results demonstrate that this particular lysine serves as a switch point for the regulation of Hsp90 functions by influencing its conformational cycle, ATPase activity, co-chaperone regulation, and client activation of yeast and human Hsp90. Incorporation of the methylated lysine via genetic code expansion specifically shows that upon modification, the conformational cycle of Hsp90 is altered. Molecular dynamics simulations including the methylated lysine suggest specific conformational changes that are propagated through Hsp90. Thus, methylation of the C-terminal lysine allows a precise allosteric tuning of Hsp90 activity via long distances.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Lysine/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Conserved Sequence , Lysine/genetics , Methylation , Molecular Dynamics Simulation , Mutation/genetics , Nucleotides/metabolism , Structure-Activity Relationship
17.
Chembiochem ; 21(13): 1861-1867, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32011787

ABSTRACT

Proteins that terminally fail to acquire their native structure are detected and degraded by cellular quality control systems. Insights into cellular protein quality control are key to a better understanding of how cells establish and maintain the integrity of their proteome and of how failures in these processes cause human disease. Here we have used genetic code expansion and fast bio-orthogonal reactions to monitor protein turnover in mammalian cells through a fluorescence-based assay. We have used immune signaling molecules (interleukins) as model substrates and shown that our approach preserves normal cellular quality control, assembly processes, and protein functionality and works for different proteins and fluorophores. We have further extended our approach to a pulse-chase type of assay that can provide kinetic insights into cellular protein behavior. Taken together, this study establishes a minimally invasive method to investigate protein turnover in cells as a key determinant of cellular homeostasis.


Subject(s)
Fluorescent Dyes/chemistry , Interleukins/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/metabolism , HEK293 Cells , Half-Life , Humans , Interleukins/chemistry , Interleukins/genetics , Kinetics , Protein Folding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
18.
Angew Chem Int Ed Engl ; 58(44): 15876-15882, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31476269

ABSTRACT

Inverse electron-demand Diels-Alder cycloadditions (iEDDAC) between tetrazines and strained alkenes/alkynes have emerged as essential tools for studying and manipulating biomolecules. A light-triggered version of iEDDAC (photo-iEDDAC) is presented that confers spatio-temporal control to bioorthogonal labeling in vitro and in cellulo. A cyclopropenone-caged dibenzoannulated bicyclo[6.1.0]nonyne probe (photo-DMBO) was designed that is unreactive towards tetrazines before light-activation, but engages in iEDDAC after irradiation at 365 nm. Aminoacyl tRNA synthetase/tRNA pairs were discovered for efficient site-specific incorporation of tetrazine-containing amino acids into proteins in living cells. In situ light activation of photo-DMBO conjugates allows labeling of tetrazine-modified proteins in living E. coli. This allows proteins in living cells to be modified in a spatio-temporally controlled manner and may be extended to photo-induced and site-specific protein labeling in animals.


Subject(s)
Bridged Bicyclo Compounds/chemistry , Cyclopropanes/chemistry , Escherichia coli Proteins/chemistry , Fluorescent Dyes/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Light , Bridged Bicyclo Compounds/chemical synthesis , Cycloaddition Reaction , Fluorescent Dyes/chemical synthesis , Molecular Structure , Photochemical Processes
19.
Biochemistry ; 58(24): 2703-2705, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31181902

Subject(s)
Alkynes , Proteins , Bacteria
20.
Chem Commun (Camb) ; 55(33): 4793-4796, 2019 Apr 18.
Article in English | MEDLINE | ID: mdl-30945708

ABSTRACT

We report the genetically encoded incorporation of phenylacetyl protected lysine (PacK) into proteins in Escherichia coli. This unnatural side-chain modification can be enzymatically removed using either penicillin G acylase (PGA) or, surprisingly, the sirtuin SrtN from Bacillus subtilis. Our approach expands the toolbox to reversibly control protein structure and function under very mild and non-denaturing conditions, as demonstrated by triggering the activity of the nonribosomal peptide synthetase GrsA.

SELECTION OF CITATIONS
SEARCH DETAIL
...