Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 473
Filter
1.
Nat Commun ; 15(1): 6568, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39095377

ABSTRACT

Developing programmable bacterial cell-cell adhesion is of significant interest due to its versatile applications. Current methods that rely on presenting cell adhesion molecules (CAMs) on bacterial surfaces are limited by the lack of a generalizable strategy to identify such molecules targeting bacterial membrane proteins in their natural states. Here, we introduce a whole-cell screening platform designed to discover CAMs targeting bacterial membrane proteins within a synthetic bacteria-displayed nanobody library. Leveraging the potency of the bacterial type IV secretion system-a contact-dependent DNA delivery nanomachine-we have established a positive feedback mechanism to selectively enrich for bacteria displaying nanobodies that target antigen-expressing cells. Our platform successfully identified functional CAMs capable of recognizing three distinct outer membrane proteins (TraN, OmpA, OmpC), demonstrating its efficacy in CAM discovery. This approach holds promise for engineering bacterial cell-cell adhesion, such as directing the antibacterial activity of programmed inhibitor cells toward target bacteria in mixed populations.


Subject(s)
Bacterial Adhesion , Cell Adhesion Molecules , Single-Domain Antibodies , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Single-Domain Antibodies/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Escherichia coli/metabolism , Bacteria/metabolism
2.
Nat Commun ; 15(1): 7228, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174501

ABSTRACT

The Wnt receptor Frizzled3 (FZD3) is important for brain axonal development and cancer progression. We report structures of FZD3 in complex with extracellular and intracellular binding nanobodies (Nb). The crystal structure of Nb8 in complex with the FZD3 cysteine-rich domain (CRD) reveals that the nanobody binds at the base of the lipid-binding groove and can compete with Wnt5a. Nb8 fused with the Dickkopf-1 C-terminal domain behaves as a FZD3-specific Wnt surrogate, activating ß-catenin signalling. The cryo-EM structure of FZD3 in complex with Nb9 reveals partially resolved density for the CRD, which exhibits positional flexibility, and a transmembrane conformation that resembles active GPCRs. Nb9 binds to the cytoplasmic region of FZD3 at the putative Dishevelled (DVL) or G protein-binding site, competes with DVL binding, and inhibits GαS coupling. In combination, our FZD3 structures with nanobody modulators map extracellular and intracellular interaction surfaces of functional, and potentially therapeutic, relevance.


Subject(s)
Frizzled Receptors , Single-Domain Antibodies , Frizzled Receptors/metabolism , Frizzled Receptors/chemistry , Humans , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Protein Binding , Crystallography, X-Ray , HEK293 Cells , Binding Sites , Cryoelectron Microscopy , Animals , Models, Molecular , Protein Domains , Dishevelled Proteins/metabolism , Dishevelled Proteins/chemistry , Dishevelled Proteins/genetics , Wnt Signaling Pathway , beta Catenin/metabolism , beta Catenin/chemistry
3.
Nat Commun ; 15(1): 6068, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025931

ABSTRACT

Neurexins are key adhesion proteins that coordinate extracellular and intracellular synaptic components. Nonetheless, the low abundance of these multidomain proteins has complicated any localization and structure-function studies. Here we combine an ALFA tag (AT)/nanobody (NbALFA) tool with classic genetics, cell biology and electrophysiology to examine the distribution and function of the Drosophila Nrx-1 in vivo. We generate full-length and ΔPDZ ALFA-tagged Nrx-1 variants and find that the PDZ binding motif is key to Nrx-1 surface expression. A PDZ binding motif provided in trans, via genetically encoded cytosolic NbALFA-PDZ chimera, fully restores the synaptic localization and function of NrxΔPDZ-AT. Using cytosolic NbALFA-mScarlet intrabody, we achieve compartment-specific detection of endogenous Nrx-1, track live Nrx-1 transport along the motor neuron axons, and demonstrate that Nrx-1 co-migrates with Rab2-positive vesicles. Our findings illustrate the versatility of the ALFA system and pave the way towards dissecting functional domains of complex proteins in vivo.


Subject(s)
Drosophila Proteins , Single-Domain Antibodies , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Single-Domain Antibodies/metabolism , Drosophila melanogaster/metabolism , Motor Neurons/metabolism , PDZ Domains , Axons/metabolism , Neural Cell Adhesion Molecules/metabolism , Neural Cell Adhesion Molecules/genetics , Protein Transport , Cell Adhesion Molecules, Neuronal
4.
Nat Commun ; 15(1): 6214, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043660

ABSTRACT

Protein-protein interactions (PPIs) are central in cell metabolism but research tools for the structural and functional characterization of these PPIs are often missing. Here we introduce broadly applicable immunization (Cross-link PPIs and immunize llamas, ChILL) and selection strategies (Display and co-selection, DisCO) for the discovery of diverse nanobodies that either stabilize or disrupt PPIs in a single experiment. We apply ChILL and DisCO to identify competitive, connective, or fully allosteric nanobodies that inhibit or facilitate the formation of the SOS1•RAS complex and modulate the nucleotide exchange rate on this pivotal GTPase in vitro as well as RAS signalling in cellulo. One of these connective nanobodies fills a cavity that was previously identified as the binding pocket for a series of therapeutic lead compounds. The long complementarity-determining region (CDR3) that penetrates this binding pocket serves as pharmacophore for extending the repertoire of potential leads.


Subject(s)
Protein Binding , SOS1 Protein , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Single-Domain Antibodies/metabolism , SOS1 Protein/metabolism , SOS1 Protein/chemistry , SOS1 Protein/genetics , SOS1 Protein/immunology , Humans , Animals , Allosteric Regulation , ras Proteins/metabolism , ras Proteins/chemistry , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Binding Sites , Camelids, New World/immunology , Immunization , Signal Transduction , Models, Molecular
5.
Nat Commun ; 15(1): 4687, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824166

ABSTRACT

Ligand-induced activation of G protein-coupled receptors (GPCRs) can initiate signaling through multiple distinct pathways with differing biological and physiological outcomes. There is intense interest in understanding how variation in GPCR ligand structure can be used to promote pathway selective signaling ("biased agonism") with the goal of promoting desirable responses and avoiding deleterious side effects. Here we present an approach in which a conventional peptide ligand for the type 1 parathyroid hormone receptor (PTHR1) is converted from an agonist which induces signaling through all relevant pathways to a compound that is highly selective for a single pathway. This is achieved not through variation in the core structure of the agonist, but rather by linking it to a nanobody tethering agent that binds with high affinity to a separate site on the receptor not involved in signal transduction. The resulting conjugate represents the most biased agonist of PTHR1 reported to date. This approach holds promise for facile generation of pathway selective ligands for other GPCRs.


Subject(s)
Receptor, Parathyroid Hormone, Type 1 , Receptors, G-Protein-Coupled , Signal Transduction , Single-Domain Antibodies , Ligands , Humans , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptor, Parathyroid Hormone, Type 1/agonists , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , HEK293 Cells , Signal Transduction/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Protein Binding , Animals , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism
6.
J Med Chem ; 67(12): 9857-9868, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38842931

ABSTRACT

Increasing research efforts focus on exploiting antibodies to inhibit the amyloid formation of neurodegenerative proteins. Nevertheless, it is challenging to discover antibodies that inhibit this process in a specific manner. Using ribosome display, we screened for synthetic single-domain antibodies, i.e., sybodies, of the P1 region of α-synuclein (residues 36-42), a protein that forms amyloid in Parkinson's disease and multiple-system atrophy. Hits were assessed for direct binding to a P1 peptide and the inhibition of amyloid formation. We discovered a sybody, named αSP1, that inhibits amyloid formation of α-synuclein at substoichiometric concentrations in a specific manner, even within highly crowded heterogeneous mixtures. Fluorescence resonance energy transfer-based binding assays and seeding experiments with and without αSP1 further demonstrate the importance of the P1 region for both primary and secondary nucleation mechanisms of amyloid assembly.


Subject(s)
Amyloid , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/antagonists & inhibitors , Humans , Amyloid/metabolism , Amyloid/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Protein Binding
7.
J Mol Biol ; 436(16): 168667, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38901640

ABSTRACT

The excessive deposition of fibrillar collagens is a hallmark of fibrosis. Collagen fibril formation requires proteolytic maturations by Procollagen N- and C-proteinases (PNPs and PCPs) to remove the N- and C-propeptides which maintain procollagens in the soluble form. Procollagen C-Proteinase Enhancer-1 (PCPE-1, a glycoprotein composed of two CUB domains and one NTR domain) is a regulatory protein that activates the C-terminal processing of procollagens by the main PCPs. It is often up-regulated in fibrotic diseases and represents a promising target for the development of novel anti-fibrotic strategies. Here, our objective was to develop the first antagonists of PCPE-1, based on the nanobody scaffold. Using both an in vivo selection through the immunization of a llama and an in vitro selection with a synthetic library, we generated 18 nanobodies directed against the CUB domains of PCPE1, which carry its enhancing activity. Among them, I5 from the immune library and H4 from the synthetic library have a high affinity for PCPE-1 and inhibit its interaction with procollagens. The crystal structure of the complex formed by PCPE-1, H4 and I5 showed that they have distinct epitopes and enabled the design of a biparatopic fusion, the diabody diab-D1. Diab-D1 has a sub-nanomolar affinity for PCPE-1 and is a potent antagonist of its activity, preventing the stimulation of procollagen cleavage in vitro. Moreover, Diab-D1 is also effective in reducing the proteolytic maturation of procollagen I in cultures of human dermal fibroblasts and hence holds great promise as a tool to modulate collagen deposition in fibrotic conditions.


Subject(s)
Proteolysis , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/immunology , Animals , Humans , Camelids, New World , Procollagen/metabolism , Procollagen/chemistry , Protein Domains , Models, Molecular , Fibrillar Collagens/metabolism , Fibrillar Collagens/chemistry , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/chemistry , Crystallography, X-Ray
8.
ACS Chem Biol ; 19(7): 1426-1432, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38941516

ABSTRACT

Chemokines are an important family of small proteins integral to leukocyte recruitment during inflammation. Dysregulation of the chemokine-chemokine receptor axis is implicated in many diseases, and both chemokines and their cognate receptors have been the targets of therapeutic development. Analysis of the antigen-binding regions of chemokine-binding nanobodies revealed a sequence motif suggestive of tyrosine sulfation. Given the well-established importance of post-translational tyrosine sulfation of receptors for chemokine affinity, it was hypothesized that the sulfation of these nanobodies may contribute to chemokine binding and selectivity. Four nanobodies (16C1, 9F1, 11B1, and 11F2) were expressed using amber codon suppression to incorporate tyrosine sulfation. The sulfated variant of 16C1 demonstrated significantly improved chemokine binding compared to the non-sulfated counterpart, while the other nanobodies displayed equipotent or reduced affinity upon sulfation. The ability of tyrosine sulfation to modulate chemokine binding, both positively and negatively, could be leveraged for chemokine-targeted sulfo-nanobody therapeutics in the future.


Subject(s)
Chemokines , Single-Domain Antibodies , Tyrosine , Tyrosine/metabolism , Tyrosine/chemistry , Tyrosine/analogs & derivatives , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Chemokines/metabolism , Chemokines/chemistry , Humans , Protein Binding , Sulfates/metabolism , Sulfates/chemistry
9.
Nucleic Acids Res ; 52(14): e66, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38932691

ABSTRACT

Transcription factors (TFs) form homo- or hetero-dimeric DNA binding complexes along with associated co-regulators that can have transcriptional repressor or activator functions. Defining the specific composition of the complexes is therefore key to understanding their biological role. Here, we utilized bimolecular fluorescence complementation (BiFC) to visualize the formation of defined TF dimers and associated co-regulators derived from the activator protein-1 (AP-1) and myocyte enhancer factor 2 (MEF2) families. Firstly, BiFC signals were observed in cells co-expressing TFs tagged with complimentary combinations of the split fluorescent protein, demonstrating the engineered formation of defined dimer complexes. Next, we applied this approach and determined that defined AP-1 dimers localized at discrete sub-nuclear locations. Subsequently, a combination of BiFC coupled with GFP binding peptide (GBP)-nanotrap allowed observation of protein-protein interactions between a co-regulator, HDAC4, and defined BiFC-MEF2 engineered dimers. To determine transactivation properties of defined TF dimers in a cellular system, the Gal4-DNA binding domain fused to GBP was utilized to assess the transcriptional properties of the BiFC-TF dimers using a generically applicable Gal4/UAS luciferase reporter gene assay system. Here, we report efficacy of a BiFC/GBP-nanobody approach that allows engineering, visualization, and functional analysis of defined TF dimers.


Subject(s)
Green Fluorescent Proteins , Humans , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Protein Binding , Transcription Factor AP-1/metabolism , Transcription Factor AP-1/genetics , MEF2 Transcription Factors/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/chemistry , Histone Deacetylases/metabolism , Protein Multimerization , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/genetics , HEK293 Cells , Protein Interaction Mapping/methods , Transcription Factors/metabolism , Transcription Factors/genetics , Transcriptional Activation , Repressor Proteins/metabolism , Repressor Proteins/genetics , Animals
10.
Lab Chip ; 24(11): 2944-2957, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38716822

ABSTRACT

In vitro display technologies such as yeast display have been instrumental in developing the selection of new antibodies, antibody fragments or nanobodies that bind to a specific target, with affinity towards the target being the main factor that influences selection outcome. However, the roles of mechanical forces are being increasingly recognized as a crucial factor in the regulation and activation of effector cell function. It would thus be of interest to isolate binders behaving optimally under the influence of mechanical forces. We developed a microfluidic assay allowing the selection of yeast displaying nanobodies through antigen-specific immobilization on a surface under controlled hydrodynamic flow. This approach enabled enrichment of model yeast mixtures using tunable antigen density and applied force. This new force-based selection method opens the possibility of selecting binders by relying on both their affinity and force resistance, with implications for the design of more efficient immunotherapeutics.


Subject(s)
Antigens , Saccharomyces cerevisiae , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/immunology , Saccharomyces cerevisiae/metabolism , Antigens/immunology , Antigens/metabolism , Microfluidic Analytical Techniques/instrumentation
11.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38816213

ABSTRACT

In cells, mitochondria undergo constant fusion and fission. An essential factor for fission is the mammalian dynamin-related protein 1 (Drp1). Dysregulation of Drp1 is associated with neurodegenerative diseases including Parkinson's, cardiovascular diseases and cancer, making Drp1 a pivotal biomarker for monitoring mitochondrial status and potential pathophysiological conditions. Here, we developed nanobodies (Nbs) as versatile binding molecules for proteomics, advanced microscopy and live cell imaging of Drp1. To specifically enrich endogenous Drp1 with interacting proteins for proteomics, we functionalized high-affinity Nbs into advanced capture matrices. Furthermore, we detected Drp1 by bivalent Nbs combined with site-directed fluorophore labelling in super-resolution STORM microscopy. For real-time imaging of Drp1, we intracellularly expressed fluorescently labelled Nbs, so-called chromobodies (Cbs). To improve the signal-to-noise ratio, we further converted Cbs into a "turnover-accelerated" format. With these imaging probes, we visualized the dynamics of endogenous Drp1 upon compound-induced mitochondrial fission in living cells. Considering the wide range of research applications, the presented Nb toolset will open up new possibilities for advanced functional studies of Drp1 in disease-relevant models.


Subject(s)
Dynamins , Mitochondria , Mitochondrial Dynamics , Single-Domain Antibodies , Dynamins/metabolism , Humans , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/immunology , Mitochondria/metabolism , Proteomics/methods , Animals , Protein Binding , HeLa Cells , Mitochondrial Proteins/metabolism
12.
Nat Commun ; 15(1): 4611, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816420

ABSTRACT

G protein-coupled receptors (GPCRs) are pivotal therapeutic targets, but their complex structure poses challenges for effective drug design. Nanobodies, or single-domain antibodies, have emerged as a promising therapeutic strategy to target GPCRs, offering advantages over traditional small molecules and antibodies. However, an incomplete understanding of the structural features enabling GPCR-nanobody interactions has limited their development. In this study, we investigate VUN701, a nanobody antagonist targeting the atypical chemokine receptor 3 (ACKR3). We determine that an extended CDR3 loop is required for ACKR3 binding. Uncommon in most nanobodies, an extended CDR3 is prevalent in GPCR-targeting nanobodies. Combining experimental and computational approaches, we map an inhibitory ACKR3-VUN701 interface and define a distinct conformational mechanism for GPCR inactivation. Our results provide insights into class A GPCR-nanobody selectivity and suggest a strategy for the development of these new therapeutic tools.


Subject(s)
Receptors, CXCR , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Humans , Receptors, CXCR/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/chemistry , HEK293 Cells , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals
13.
J Am Chem Soc ; 146(23): 16173-16183, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38819260

ABSTRACT

Genetically encoding a proximal reactive warhead into the protein binder/drug has emerged as an efficient strategy for covalently binding to protein targets, enabling broad applications. To expand the reactivity scope for targeting the diverse natural residues under physiological conditions, the development of a genetically encoded reactive warhead with excellent stability and broad reactivity is highly desired. Herein, we reported the genetic encoding of epoxide-containing tyrosine (EPOY) for developing covalent protein drugs. Our study demonstrates that EPOY, when incorporated into a nanobody (KN035), can cross-link with different side chains (mutations) at the same position of PD-L1 protein. Significantly, a single genetically encoded reactive warhead that is capable of covalent and site-specific targeting to 10 different nucleophilic residues was achieved for the first time. This would largely expand the scope of covalent warhead and inspire the development of covalent warheads for both small-molecule drugs and protein drugs. Furthermore, we incorporate the EPOY into a designed ankyrin repeat protein (DarpinK13) to create the covalent binders of KRAS. This covalent KRAS binder holds the potential to achieve pan-covalent targeting of KRAS based on the structural similarity among all oncogenic KRAS mutants while avoiding off-target binding to NRAS/HRAS through a covalent interaction with KRAS-specific residues (H95 and E107). We envision that covalently targeting to H95 will be a promising strategy for the development of covalent pan-KRAS inhibitors in the future.


Subject(s)
Epoxy Compounds , Humans , Epoxy Compounds/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Tyrosine/chemistry , B7-H1 Antigen/chemistry , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism
14.
Elife ; 122024 May 28.
Article in English | MEDLINE | ID: mdl-38805257

ABSTRACT

Mycobacterium tuberculosis (Mtb) is known to survive within macrophages by compromising the integrity of the phagosomal compartment in which it resides. This activity primarily relies on the ESX-1 secretion system, predominantly involving the protein duo ESAT-6 and CFP-10. CFP-10 likely acts as a chaperone, while ESAT-6 likely disrupts phagosomal membrane stability via a largely unknown mechanism. we employ a series of biochemical analyses, protein modeling techniques, and a novel ESAT-6-specific nanobody to gain insight into the ESAT-6's mode of action. First, we measure the binding kinetics of the tight 1:1 complex formed by ESAT-6 and CFP-10 at neutral pH. Subsequently, we demonstrate a rapid self-association of ESAT-6 into large complexes under acidic conditions, leading to the identification of a stable tetrameric ESAT-6 species. Using molecular dynamics simulations, we pinpoint the most probable interaction interface. Furthermore, we show that cytoplasmic expression of an anti-ESAT-6 nanobody blocks Mtb replication, thereby underlining the pivotal role of ESAT-6 in intracellular survival. Together, these data suggest that ESAT-6 acts by a pH-dependent mechanism to establish two-way communication between the cytoplasm and the Mtb-containing phagosome.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Macrophages , Mycobacterium tuberculosis , Phagosomes , Single-Domain Antibodies , Humans , Antigens, Bacterial/metabolism , Antigens, Bacterial/immunology , Bacterial Proteins/metabolism , Hydrogen-Ion Concentration , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Molecular Dynamics Simulation , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Phagosomes/metabolism , Single-Domain Antibodies/metabolism
15.
PLoS One ; 19(5): e0303839, 2024.
Article in English | MEDLINE | ID: mdl-38758765

ABSTRACT

The interaction between SARS-CoV-2 non-structural protein Nsp9 and the nanobody 2NSP90 was investigated by NMR spectroscopy using the paramagnetic perturbation methodology PENELOP (Paramagnetic Equilibrium vs Nonequilibrium magnetization Enhancement or LOss Perturbation). The Nsp9 monomer is an essential component of the replication and transcription complex (RTC) that reproduces the viral gRNA for subsequent propagation. Therefore preventing Nsp9 recruitment in RTC would represent an efficient antiviral strategy that could be applied to different coronaviruses, given the Nsp9 relative invariance. The NMR results were consistent with a previous characterization suggesting a 4:4 Nsp9-to-nanobody stoichiometry with the occurrence of two epitope pairs on each of the Nsp9 units that establish the inter-dimer contacts of Nsp9 tetramer. The oligomerization state of Nsp9 was also analyzed by molecular dynamics simulations and both dimers and tetramers resulted plausible. A different distribution of the mapped epitopes on the tetramer surface with respect to the former 4:4 complex could also be possible, as well as different stoichiometries of the Nsp9-nanobody assemblies such as the 2:2 stoichiometry suggested by the recent crystal structure of the Nsp9 complex with 2NSP23 (PDB ID: 8dqu), a nanobody exhibiting essentially the same affinity as 2NSP90. The experimental NMR evidence, however, ruled out the occurrence in liquid state of the relevant Nsp9 conformational change observed in the same crystal structure.


Subject(s)
Epitopes , Molecular Dynamics Simulation , SARS-CoV-2 , Single-Domain Antibodies , Viral Nonstructural Proteins , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Single-Domain Antibodies/metabolism , SARS-CoV-2/immunology , Epitopes/immunology , Epitopes/chemistry , Humans , Magnetic Resonance Spectroscopy , Protein Binding , Protein Multimerization , COVID-19/immunology , COVID-19/virology , RNA-Binding Proteins
16.
Nat Commun ; 15(1): 4173, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755204

ABSTRACT

Potassium channels of the Two-Pore Domain (K2P) subfamily, KCNK1-KCNK18, play crucial roles in controlling the electrical activity of many different cell types and represent attractive therapeutic targets. However, the identification of highly selective small molecule drugs against these channels has been challenging due to the high degree of structural and functional conservation that exists not only between K2P channels, but across the whole K+ channel superfamily. To address the issue of selectivity, here we generate camelid antibody fragments (nanobodies) against the TREK-2 (KCNK10) K2P K+ channel and identify selective binders including several that directly modulate channel activity. X-ray crystallography and CryoEM data of these nanobodies in complex with TREK-2 also reveal insights into their mechanisms of activation and inhibition via binding to the extracellular loops and Cap domain, as well as their suitability for immunodetection. These structures facilitate design of a biparatropic inhibitory nanobody with markedly improved sensitivity. Together, these results provide important insights into TREK channel gating and provide an alternative, more selective approach to modulation of K2P channel activity via their extracellular domains.


Subject(s)
Potassium Channels, Tandem Pore Domain , Single-Domain Antibodies , Potassium Channels, Tandem Pore Domain/metabolism , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Humans , Crystallography, X-Ray , Animals , Cryoelectron Microscopy , HEK293 Cells , Models, Molecular
17.
Elife ; 132024 Apr 26.
Article in English | MEDLINE | ID: mdl-38666771

ABSTRACT

Roco proteins entered the limelight after mutations in human LRRK2 were identified as a major cause of familial Parkinson's disease. LRRK2 is a large and complex protein combining a GTPase and protein kinase activity, and disease mutations increase the kinase activity, while presumably decreasing the GTPase activity. Although a cross-communication between both catalytic activities has been suggested, the underlying mechanisms and the regulatory role of the GTPase domain remain unknown. Several structures of LRRK2 have been reported, but structures of Roco proteins in their activated GTP-bound state are lacking. Here, we use single-particle cryo-electron microscopy to solve the structure of a bacterial Roco protein (CtRoco) in its GTP-bound state, aided by two conformation-specific nanobodies: NbRoco1 and NbRoco2. This structure presents CtRoco in an active monomeric state, featuring a very large GTP-induced conformational change using the LRR-Roc linker as a hinge. Furthermore, this structure shows how NbRoco1 and NbRoco2 collaborate to activate CtRoco in an allosteric way. Altogether, our data provide important new insights into the activation mechanism of Roco proteins, with relevance to LRRK2 regulation, and suggest new routes for the allosteric modulation of their GTPase activity.


Subject(s)
Cryoelectron Microscopy , Guanosine Triphosphate , Single-Domain Antibodies , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/chemistry , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/chemistry , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Protein Conformation , Allosteric Regulation , Models, Molecular , Protein Multimerization , Humans
18.
Biomolecules ; 14(4)2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38672440

ABSTRACT

This study assessed the suitability of the complementarity-determining region 2 (CDR2) of the nanobody (Nb) as a template for the derivation of nanobody-derived peptides (NDPs) targeting active-state ß2-adrenergic receptor (ß2AR) conformation. Sequences of conformationally selective Nbs favoring the agonist-occupied ß2AR were initially analyzed by the informational spectrum method (ISM). The derived NDPs in complex with ß2AR were subjected to protein-peptide docking, molecular dynamics (MD) simulations, and metadynamics-based free-energy binding calculations. Computational analyses identified a 25-amino-acid-long CDR2-NDP of Nb71, designated P4, which exhibited the following binding free-energy for the formation of the ß2AR:P4 complex (ΔG = -6.8 ± 0.8 kcal/mol or a Ki = 16.5 µM at 310 K) and mapped the ß2AR:P4 amino acid interaction network. In vitro characterization showed that P4 (i) can cross the plasma membrane, (ii) reduces the maximum isoproterenol-induced cAMP level by approximately 40% and the isoproterenol potency by up to 20-fold at micromolar concentration, (iii) has a very low affinity to interact with unstimulated ß2AR in the cAMP assay, and (iv) cannot reduce the efficacy and potency of the isoproterenol-mediated ß2AR/ß-arrestin-2 interaction in the BRET2-based recruitment assay. In summary, the CDR2-NDP, P4, binds preferentially to agonist-activated ß2AR and disrupts Gαs-mediated signaling.


Subject(s)
Peptides , Receptors, Adrenergic, beta-2 , Single-Domain Antibodies , Humans , Amino Acid Sequence , Complementarity Determining Regions/chemistry , Cyclic AMP/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism , Protein Binding , Protein Conformation , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-2/chemistry , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/metabolism
19.
PLoS One ; 19(4): e0300453, 2024.
Article in English | MEDLINE | ID: mdl-38683783

ABSTRACT

The activity-regulated cytoskeleton-associated protein (Arc) is a complex regulator of synaptic plasticity in glutamatergic neurons. Understanding its molecular function is key to elucidate the neurobiology of memory and learning, stress regulation, and multiple neurological and psychiatric diseases. The recent development of anti-Arc nanobodies has promoted the characterization of the molecular structure and function of Arc. This study aimed to validate two anti-Arc nanobodies, E5 and H11, as selective modulators of the human Arc N-lobe (Arc-NL), a domain that mediates several molecular functions of Arc through its peptide ligand binding site. The structural characteristics of recombinant Arc-NL-nanobody complexes were solved at atomic resolution using X-ray crystallography. Both anti-Arc nanobodies bind specifically to the multi-peptide binding site of Arc-NL. Isothermal titration calorimetry showed that the Arc-NL-nanobody interactions occur at nanomolar affinity, and that the nanobodies can displace a TARPγ2-derived peptide from the binding site. Thus, both anti-Arc-NL nanobodies could be used as competitive inhibitors of endogenous Arc ligands. Differences in the CDR3 loops between the two nanobodies indicate that the spectrum of short linear motifs recognized by the Arc-NL should be expanded. We provide a robust biochemical background to support the use of anti-Arc nanobodies in attempts to target Arc-dependent synaptic plasticity. Function-blocking anti-Arc nanobodies could eventually help unravel the complex neurobiology of synaptic plasticity and allow to develop diagnostic and treatment tools.


Subject(s)
Cytoskeletal Proteins , Nerve Tissue Proteins , Single-Domain Antibodies , Humans , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Single-Domain Antibodies/metabolism , Binding Sites , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/immunology , Ligands , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/immunology , Crystallography, X-Ray , Protein Binding , Models, Molecular , Amino Acid Sequence
20.
Microb Cell Fact ; 23(1): 124, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689251

ABSTRACT

BACKGROUND: Having a simple and fast dividing organism capable of producing and exposing at its surface or secreting functional complex biomolecules with disulphide bridges is of great interest. The mycoplasma bacterial genus offers a set of relevant properties that make it an interesting chassis for such purposes, the main one being the absence of a cell wall. However, due to their slow growth, they have rarely been considered as a potential platform in this respect. This notion may be challenged with the recent discovery of Mycoplasma feriruminatoris, a species with a dividing time close to that of common microbial workhorses. So far, no tools for heterologous protein expression nor secretion have been described for it. RESULTS: The work presented here develops the fast-dividing M. feriruminatoris as a tool for secreting functional biomolecules of therapeutic interest that could be used for screening functional mutants as well as potentially for protein-protein interactions. Based on RNAseq, quantitative proteomics and promoter sequence comparison we have rationally designed optimal promoter sequences. Then, using in silico analysis, we have identified putative secretion signals that we validated using a luminescent reporter. The potential of the resulting secretion cassette has been shown with set of active clinically relevant proteins (interleukins and nanobodies). CONCLUSIONS: We have engineered Mycoplasma feriruminatoris for producing and secreting functional proteins of medical interest.


Subject(s)
Bacterial Proteins , Mycoplasma , Mycoplasma/metabolism , Mycoplasma/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Promoter Regions, Genetic , Proteomics , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/genetics
SELECTION OF CITATIONS
SEARCH DETAIL