Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.618
Filtrar
1.
Elife ; 132024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39302337

RESUMO

Glycans play an important role in modulating the interactions between natural killer cells and antibodies to fight pathogens and harmful cells.


Assuntos
Células Matadoras Naturais , Polissacarídeos , Polissacarídeos/metabolismo , Polissacarídeos/imunologia , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/metabolismo
2.
Cells ; 13(17)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39273046

RESUMO

One of the important genes for eyespot development in butterfly wings is Distal-less. Its function has been evaluated via several methods, including CRISPR/Cas9 genome editing. However, functional inhibition may be performed at the right time at the right place using a different method. Here, we used a novel protein delivery method for pupal wing tissues in vivo to inactivate a target protein, Distal-less, with a polyclonal anti-Distal-less antibody using the blue pansy butterfly Junonia orithya. We first demonstrated that various antibodies including the anti-Distal-less antibody were delivered to wing epithelial cells in vivo in this species. Treatment with the anti-Distal-less antibody reduced eyespot size, confirming the positive role of Distal-less in eyespot development. The treatment eliminated or deformed a parafocal element, suggesting a positive role of Distal-less in the development of the parafocal element. This result also suggested the integrity of an eyespot and its corresponding parafocal element as the border symmetry system. Taken together, these findings demonstrate that the antibody-mediated protein knockdown method is a useful tool for functional assays of proteins, such as Distal-less, expressed in pupal wing tissues, and that Distal-less functions for eyespots and parafocal elements in butterfly wing color pattern development.


Assuntos
Anticorpos , Borboletas , Proteínas de Insetos , Asas de Animais , Animais , Borboletas/metabolismo , Borboletas/genética , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Anticorpos/metabolismo , Proteínas de Insetos/metabolismo , Proteínas de Insetos/genética , Pigmentação/genética , Técnicas de Silenciamento de Genes
3.
Wiley Interdiscip Rev RNA ; 15(4): e1869, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39187256

RESUMO

RNA structure is crucial to a wide range of cellular processes. The intimate relationship between macromolecular structure and function necessitates the determination of high-resolution structures of functional RNA molecules. X-ray crystallography is the predominant technique used for macromolecular structure determination; however, solving RNA structures has been more challenging than their protein counterparts, as reflected in their poor representation in the Protein Data Bank (<1%). Antibody-assisted RNA crystallography is a relatively new technique that promises to accelerate RNA structure determination by employing synthetic antibodies (Fabs) as crystallization chaperones that are specifically raised against target RNAs. Antibody chaperones facilitate the formation of ordered crystal lattices by minimizing RNA flexibility and replacing unfavorable RNA-RNA contacts with contacts between chaperone molecules. Atomic coordinates of these antibody fragments can also be used as search models to obtain phase information during structure determination. Antibody-assisted RNA crystallography has enabled the structure determination of 15 unique RNA targets, including 11 in the last 6 years. In this review, I cover the historical development of antibody fragments as crystallization chaperones and their application to diverse RNA targets. I discuss how the first structures of antibody-RNA complexes informed the design of second-generation antibodies and led to the development of portable crystallization modules that have greatly reduced the uncertainties associated with RNA crystallography. Finally, I outline unexplored avenues that can increase the impact of this technology in structural biology research and discuss potential applications of antibodies as affinity reagents for interrogating RNA biology outside of their use in crystallography. This article is categorized under: RNA Structure and Dynamics > RNA Structure, Dynamics and Chemistry RNA Interactions with Proteins and Other Molecules > Protein-RNA Recognition RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes.


Assuntos
RNA , RNA/química , Cristalografia por Raios X , Anticorpos/química , Anticorpos/metabolismo , Conformação de Ácido Nucleico , Humanos
4.
Elife ; 132024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39206942

RESUMO

Immunofluorescence localises proteins via fluorophore-labelled antibodies. However, some proteins evade detection due to antibody-accessibility issues or because they are naturally low abundant or antigen density is reduced by the imaging method. Here, we show that the fusion of the target protein to the biotin ligase TurboID and subsequent detection of biotinylation by fluorescent streptavidin offers an 'all in one' solution to these restrictions. For all proteins tested, the streptavidin signal was significantly stronger than an antibody signal, markedly improving the sensitivity of expansion microscopy and correlative light and electron microscopy. Importantly, proteins within phase-separated regions, such as the central channel of the nuclear pores, the nucleolus, or RNA granules, were readily detected with streptavidin, while most antibodies failed. When TurboID is used in tandem with an HA epitope tag, co-probing with streptavidin and anti-HA can map antibody-accessibility and we created such a map for the trypanosome nuclear pore. Lastly, we show that streptavidin imaging resolves dynamic, temporally, and spatially distinct sub-complexes and, in specific cases, reveals a history of dynamic protein interaction. In conclusion, streptavidin imaging has major advantages for the detection of lowly abundant or inaccessible proteins and in addition, provides information on protein interactions and biophysical environment.


Assuntos
Estreptavidina , Estreptavidina/química , Estreptavidina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Anticorpos/metabolismo , Humanos , Biotinilação , Microscopia de Fluorescência/métodos , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo
5.
J Med Chem ; 67(16): 14110-14124, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39051153

RESUMO

C5a is an integral glycoprotein of the complement system that plays an important role in inflammation and immunity. The physiological concentration of C5a is observed to be elevated under various immunoinflammatory pathophysiological conditions in humans. The pathophysiology of C5a is linked to the "two-site" protein-protein interactions (PPIs) with two genomically related receptors, such as C5aR1 and C5aR2. Therefore, pharmacophores that can potentially block the PPIs between C5a-C5aR1 and C5a-C5aR2 have tremendous potential for development as future therapeutics. Notably, the FDA has already approved antibodies that target the precursors of C5a (Eculizumab, 148 kDa) and C5a (Vilobelimab, 149 kDa) for marketing as complement-targeted therapeutics. In this context, the current study reports the structural characterization of a pair of synthetic designer antibody-like peptides (DePA and DePA1; ≤3.8 kDa) that bind to hotspot regions on C5a and also demonstrates potential traits to neutralize the function of C5a under pathophysiological conditions.


Assuntos
Complemento C5a , Peptídeos , Receptor da Anafilatoxina C5a , Transdução de Sinais , Humanos , Receptor da Anafilatoxina C5a/metabolismo , Receptor da Anafilatoxina C5a/química , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Complemento C5a/metabolismo , Complemento C5a/química , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ligação Proteica , Anticorpos/química , Anticorpos/metabolismo , Anticorpos/imunologia , Desenho de Fármacos
6.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38960407

RESUMO

The optimization of therapeutic antibodies through traditional techniques, such as candidate screening via hybridoma or phage display, is resource-intensive and time-consuming. In recent years, computational and artificial intelligence-based methods have been actively developed to accelerate and improve the development of therapeutic antibodies. In this study, we developed an end-to-end sequence-based deep learning model, termed AttABseq, for the predictions of the antigen-antibody binding affinity changes connected with antibody mutations. AttABseq is a highly efficient and generic attention-based model by utilizing diverse antigen-antibody complex sequences as the input to predict the binding affinity changes of residue mutations. The assessment on the three benchmark datasets illustrates that AttABseq is 120% more accurate than other sequence-based models in terms of the Pearson correlation coefficient between the predicted and experimental binding affinity changes. Moreover, AttABseq also either outperforms or competes favorably with the structure-based approaches. Furthermore, AttABseq consistently demonstrates robust predictive capabilities across a diverse array of conditions, underscoring its remarkable capacity for generalization across a wide spectrum of antigen-antibody complexes. It imposes no constraints on the quantity of altered residues, rendering it particularly applicable in scenarios where crystallographic structures remain unavailable. The attention-based interpretability analysis indicates that the causal effects of point mutations on antibody-antigen binding affinity changes can be visualized at the residue level, which might assist automated antibody sequence optimization. We believe that AttABseq provides a fiercely competitive answer to therapeutic antibody optimization.


Assuntos
Complexo Antígeno-Anticorpo , Aprendizado Profundo , Complexo Antígeno-Anticorpo/química , Antígenos/química , Antígenos/genética , Antígenos/metabolismo , Antígenos/imunologia , Afinidade de Anticorpos , Sequência de Aminoácidos , Biologia Computacional/métodos , Humanos , Mutação , Anticorpos/química , Anticorpos/imunologia , Anticorpos/genética , Anticorpos/metabolismo
7.
ACS Synth Biol ; 13(8): 2629-2634, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39052526

RESUMO

We recently developed "autonomous hypermutation yeast surface display" (AHEAD), a technology that enables the rapid generation of potent and specific antibodies in yeast. AHEAD pairs yeast surface display with an error-prone orthogonal DNA replication system (OrthoRep) to continuously and rapidly mutate surface-displayed antibodies, thereby enabling enrichment for stronger binding variants through repeated rounds of cell growth and fluorescence-activated cell sorting. AHEAD currently utilizes a standard galactose induction system to drive the selective display of antibodies on the yeast surface. However, achieving maximal display levels can require up to 48 h of induction. Here we report an updated version of the AHEAD platform that utilizes a synthetic ß-estradiol-induced gene expression system to regulate the surface display of antibodies and find that induction is notably faster in achieving surface display for both our AHEAD system and traditional yeast surface display from nuclear plasmids that do not hypermutate. The updated AHEAD platform was fully functional in repeated rounds of evolution to drive the rapid evolution of antibodies.


Assuntos
Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Replicação do DNA , Técnicas de Visualização da Superfície Celular/métodos , Evolução Molecular Direcionada/métodos , Plasmídeos/genética , Anticorpos/metabolismo , Estradiol/farmacologia
8.
Sci Rep ; 14(1): 15992, 2024 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-38987432

RESUMO

Aquaporins (AQPs) are a family of water permeable channels expressed on the plasma membrane with AQP5 being the major channel expressed in several human tissues including salivary and lacrimal glands. Anti-AQP5 autoantibodies have been observed in patients with Sjögren's syndrome who are characterised by dryness of both salivary and lacrimal glands, and they have been implicated in the underlying mechanisms of glandular dysfunction. AQP5 is formed by six transmembrane helices linked with three extracellular and two intracellular loops. Develop antibodies against membrane protein extracellular loops can be a challenge due to the difficulty in maintaining these proteins as recombinant in their native form. Therefore, in this work we aimed to generate an efficient stable-transfected cell line overexpressing human AQP5 (CHO-K1/AQP5) to perform primarily cell-based phage display biopanning experiments to develop new potential recombinant antibodies targeting AQP5. We also showed that the new CHO-K1/AQP5 cell line can be used to study molecular mechanisms of AQP5 sub-cellular trafficking making these cells a useful tool for functional studies.


Assuntos
Aquaporina 5 , Cricetulus , Aquaporina 5/metabolismo , Aquaporina 5/genética , Células CHO , Humanos , Animais , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Anticorpos/metabolismo , Biblioteca de Peptídeos
9.
Sci Signal ; 17(844): eado5279, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38980923

RESUMO

In this issue of Science Signaling, Jackson et al. present a new antibody strategy to-quite literally-strap transforming growth factor-ß1 (TGF-ß1) to latent complexes in the extracellular matrix. The antibody has no effect on latent TGF-ß1 presented on the surface of immune cells and thus allows targeting of the detrimental effects of TGF-ß1 in fibrosis without affecting its beneficial immune-suppressing activities.


Assuntos
Matriz Extracelular , Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/imunologia , Humanos , Matriz Extracelular/metabolismo , Matriz Extracelular/imunologia , Animais , Fibrose , Anticorpos/imunologia , Anticorpos/metabolismo , Camundongos
10.
Biosystems ; 243: 105264, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38964652

RESUMO

Computational analysis of paratope-epitope interactions between antibodies and their corresponding antigens can facilitate our understanding of the molecular mechanism underlying humoral immunity and boost the design of new therapeutics for many diseases. The recent breakthrough in artificial intelligence has made it possible to predict protein-protein interactions and model their structures. Unfortunately, detecting antigen-binding sites associated with a specific antibody is still a challenging problem. To tackle this challenge, we implemented a deep learning model to characterize interaction patterns between antibodies and their corresponding antigens. With high accuracy, our model can distinguish between antibody-antigen complexes and other types of protein-protein complexes. More intriguingly, we can identify antigens from other common protein binding regions with an accuracy of higher than 70% even if we only have the epitope information. This indicates that antigens have distinct features on their surface that antibodies can recognize. Additionally, our model was unable to predict the partnerships between antibodies and their particular antigens. This result suggests that one antigen may be targeted by more than one antibody and that antibodies may bind to previously unidentified proteins. Taken together, our results support the precision of antibody-antigen interactions while also suggesting positive future progress in the prediction of specific pairing.


Assuntos
Anticorpos , Antígenos , Aprendizado de Máquina , Antígenos/imunologia , Anticorpos/imunologia , Anticorpos/química , Anticorpos/metabolismo , Humanos , Ligação Proteica , Epitopos/imunologia , Complexo Antígeno-Anticorpo/imunologia , Complexo Antígeno-Anticorpo/química , Biologia Computacional/métodos , Sítios de Ligação , Aprendizado Profundo , Sítios de Ligação de Anticorpos
11.
Dev Comp Immunol ; 159: 105228, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38997096

RESUMO

Leukocyte immune-type receptors (LITRs) belong to a large family of teleost immunoregulatory receptors that share phylogenetic and syntenic relationships with mammalian Fc receptor-like molecules (FCRLs). Recently, several putative stimulatory Carassius auratus (Ca)-LITR transcripts, including CaLITR3, have been identified in goldfish. CaLITR3 has four extracellular immunoglobulin-like (Ig-like) domains, a transmembrane domain containing a positively charged histidine residue, and a short cytoplasmic tail region. Additionally, the calitr3 transcript is highly expressed by goldfish primary kidney neutrophils (PKNs) and macrophages (PKMs). To further investigate the immunoregulatory potential of CaLITR3 in goldfish myeloid cells, we developed and characterized a CaLITR3-epitope-specific polyclonal antibody (anti-CaL3.D1 pAb). We show that the anti-CaL3.D1 pAb stains various hematopoietic cell types within the goldfish kidney, as well as in PKNs and PKMs. Moreover, cross-linking of the anti-CaL3.D1-pAb on PKN membranes induces phosphorylation of p38 and ERK1/2, critical components of the MAPK pathway involved in controlling a wide variety of innate immune effector responses such as NETosis, respiratory burst, and cytokine release. These findings support the stimulatory potential of CaLITR3 proteins as activators of fish granulocytes and pave the way for a more in-depth examination of the immunoregulatory functions of CaLITRs in goldfish myeloid cells.


Assuntos
Proteínas de Peixes , Carpa Dourada , Rim , Sistema de Sinalização das MAP Quinases , Neutrófilos , Receptores Imunológicos , Animais , Carpa Dourada/imunologia , Proteínas de Peixes/metabolismo , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Neutrófilos/imunologia , Rim/imunologia , Rim/citologia , Sistema de Sinalização das MAP Quinases/imunologia , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Anticorpos/imunologia , Anticorpos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Células Cultivadas , Leucócitos/imunologia , Leucócitos/metabolismo
12.
Nucleic Acids Res ; 52(14): 8052-8062, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-38908025

RESUMO

i-Motifs (iMs) are non-canonical, four-stranded secondary structures formed by stacking of hemi-protonated CH+·C base pairs in cytosine-rich DNA sequences, predominantly at pH < 7. The presence of iM structures in cells was a matter of debate until the recent development of iM-specific antibody, iMab, which was instrumental for several studies that suggested the existence of iMs in live cells and their putative biological roles. We assessed the interaction of iMab with cytosine-rich oligonucleotides by biolayer interferometry (BLI), pull-down assay and bulk-FRET experiments. Our results suggest that binding of iMab to DNA oligonucleotides is governed by the presence of runs of at least two consecutive cytosines and is generally increased in acidic conditions, irrespectively of the capacity of the sequence to adopt, or not, an iM structure. Moreover, the results of the bulk-FRET assay indicate that interaction with iMab results in unfolding of iM structures even in acidic conditions, similarly to what has been observed with hnRNP K, well-studied single-stranded DNA binding protein. Taken together, our results strongly suggest that iMab actually binds to blocks of 2-3 cytosines in single-stranded DNA, and call for more careful interpretation of results obtained with this antibody.


Assuntos
Citosina , DNA de Cadeia Simples , Motivos de Nucleotídeos , Citosina/química , Citosina/metabolismo , DNA de Cadeia Simples/química , DNA de Cadeia Simples/metabolismo , Conformação de Ácido Nucleico , Ligação Proteica , Anticorpos/química , Anticorpos/metabolismo , DNA/química , DNA/metabolismo , Sequência de Bases
13.
Protein J ; 43(3): 405-424, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38724751

RESUMO

As the demand for immunotherapy to treat and manage cancers, infectious diseases and other disorders grows, a comprehensive understanding of amino acids and their intricate role in antibody engineering has become a prime requirement. Naturally produced antibodies may not have the most suitable amino acids at the complementarity determining regions (CDR) and framework regions, for therapeutic purposes. Therefore, to enhance the binding affinity and therapeutic properties of an antibody, the specific impact of certain amino acids on the antibody's architecture must be thoroughly studied. In antibody engineering, it is crucial to identify the key amino acid residues that significantly contribute to improving antibody properties. Therapeutic antibodies with higher binding affinity and improved functionality can be achieved through modifications or substitutions with highly suitable amino acid residues. Here, we have indicated the frequency of amino acids and their association with the binding free energy in CDRs. The review also analyzes the experimental outcome of two studies that reveal the frequency of amino acids in CDRs and provides their significant correlation between the outcomes. Additionally, it discusses the various bond interactions within the antibody structure and antigen binding. A detailed understanding of these amino acid properties should assist in the analysis of antibody sequences and structures needed for designing and enhancing the overall performance of therapeutic antibodies.


Assuntos
Aminoácidos , Regiões Determinantes de Complementaridade , Engenharia de Proteínas , Aminoácidos/química , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/genética , Humanos , Engenharia de Proteínas/métodos , Anticorpos/química , Anticorpos/imunologia , Anticorpos/metabolismo , Afinidade de Anticorpos , Animais
14.
Nature ; 630(8016): 493-500, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38718835

RESUMO

The introduction of AlphaFold 21 has spurred a revolution in modelling the structure of proteins and their interactions, enabling a huge range of applications in protein modelling and design2-6. Here we describe our AlphaFold 3 model with a substantially updated diffusion-based architecture that is capable of predicting the joint structure of complexes including proteins, nucleic acids, small molecules, ions and modified residues. The new AlphaFold model demonstrates substantially improved accuracy over many previous specialized tools: far greater accuracy for protein-ligand interactions compared with state-of-the-art docking tools, much higher accuracy for protein-nucleic acid interactions compared with nucleic-acid-specific predictors and substantially higher antibody-antigen prediction accuracy compared with AlphaFold-Multimer v.2.37,8. Together, these results show that high-accuracy modelling across biomolecular space is possible within a single unified deep-learning framework.


Assuntos
Aprendizado Profundo , Ligantes , Modelos Moleculares , Proteínas , Software , Humanos , Anticorpos/química , Anticorpos/metabolismo , Antígenos/metabolismo , Antígenos/química , Aprendizado Profundo/normas , Íons/química , Íons/metabolismo , Simulação de Acoplamento Molecular , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas/química , Proteínas/metabolismo , Reprodutibilidade dos Testes , Software/normas
15.
J Microbiol Biotechnol ; 34(6): 1222-1228, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38783697

RESUMO

Protein-specific antibodies are essential for various aspects of protein research, including detection, purification, and characterization. When specific antibodies are unavailable, protein tagging is a useful alternative. Small epitope tags, typically less than 10 amino acids, are widely used in protein research due to the simple modification through PCR and reduced impact on the target protein's function compared to larger tags. The 2B8 epitope tag (RDPLPFFPP), reported by us in a previous study, has high specificity and sensitivity to the corresponding antibody. However, when attached to the C-terminus of the target protein in immunoprecipitation experiments, we observed a decrease in detection signal with reduced immunity and low protein recovery. This phenomenon was not unique to 2B8 and was also observed with the commercially available Myc tag. Our study revealed that C-terminal tagging of small epitope tags requires the addition of more than one extra amino acid to enhance (restore) antibody immunities. Moreover, among the amino acids we tested, serine was the best for the 2B8 tag. Our findings demonstrated that the interaction between a small epitope and a corresponding paratope of an antibody requires an extra amino acid at the C-terminus of the epitope. This result is important for researchers planning studies on target proteins using small epitope tags.


Assuntos
Aminoácidos , Epitopos , Epitopos/imunologia , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Camundongos , Imunoprecipitação , Formação de Anticorpos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/química
16.
Histochem Cell Biol ; 162(1-2): 41-52, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38762823

RESUMO

During development and differentiation, histone modifications dynamically change locally and globally, associated with transcriptional regulation, DNA replication and repair, and chromosome condensation. The level of histone H4 Lys20 monomethylation (H4K20me1) increases during the G2 to M phases of the cell cycle and is enriched in facultative heterochromatin, such as inactive X chromosomes in cycling cells. To track the dynamic changes of H4K20me1 in living cells, we have developed a genetically encoded modification-specific intracellular antibody (mintbody) probe that specifically binds to the modification. Here, we report the generation of knock-in mice in which the coding sequence of the mCherry-tagged version of the H4K20me1-mintbody is inserted into the Rosa26 locus. The knock-in mice, which ubiquitously expressed the H4K20me1-mintbody, developed normally and were fertile, indicating that the expression of the probe does not disturb the cell growth, development, or differentiation. Various tissues isolated from the knock-in mice exhibited nuclear fluorescence without the need for fixation. The H4K20me1-mintbody was enriched in inactive X chromosomes in developing embryos and in XY bodies during spermatogenesis. The knock-in mice will be useful for the histochemical analysis of H4K20me1 in any cell types.


Assuntos
Técnicas de Introdução de Genes , Histonas , Proteínas Luminescentes , Animais , Camundongos , Histonas/metabolismo , Proteínas Luminescentes/metabolismo , Proteínas Luminescentes/genética , Anticorpos/metabolismo , Proteína Vermelha Fluorescente , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
17.
Elife ; 132024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38717135

RESUMO

Eukaryotic chromatin is organized into functional domains, that are characterized by distinct proteomic compositions and specific nuclear positions. In contrast to cellular organelles surrounded by lipid membranes, the composition of distinct chromatin domains is rather ill described and highly dynamic. To gain molecular insight into these domains and explore their composition, we developed an antibody-based proximity biotinylation method targeting the RNA and proteins constituents. The method that we termed antibody-mediated proximity labelling coupled to mass spectrometry (AMPL-MS) does not require the expression of fusion proteins and therefore constitutes a versatile and very sensitive method to characterize the composition of chromatin domains based on specific signature proteins or histone modifications. To demonstrate the utility of our approach we used AMPL-MS to characterize the molecular features of the chromocenter as well as the chromosome territory containing the hyperactive X chromosome in Drosophila. This analysis identified a number of known RNA-binding proteins in proximity of the hyperactive X and the centromere, supporting the accuracy of our method. In addition, it enabled us to characterize the role of RNA in the formation of these nuclear bodies. Furthermore, our method identified a new set of RNA molecules associated with the Drosophila centromere. Characterization of these novel molecules suggested the formation of R-loops in centromeres, which we validated using a novel probe for R-loops in Drosophila. Taken together, AMPL-MS improves the selectivity and specificity of proximity ligation allowing for novel discoveries of weak protein-RNA interactions in biologically diverse domains.


Assuntos
Cromatina , Espectrometria de Massas , RNA , Animais , Cromatina/metabolismo , Cromatina/química , RNA/metabolismo , RNA/química , Espectrometria de Massas/métodos , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Biotinilação , Centrômero/metabolismo , Anticorpos/metabolismo , Anticorpos/química , Cromossomo X/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/química , Coloração e Rotulagem/métodos
18.
Biochem J ; 481(10): 643-651, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38683688

RESUMO

GCN5L1, also known as BLOC1S1 and BLOS1, is a small intracellular protein involved in many key biological processes. Over the last decade, GCN5L1 has been implicated in the regulation of protein lysine acetylation, energy metabolism, endo-lysosomal function, and cellular immune pathways. An increasing number of published papers have used commercially-available reagents to interrogate GCN5L1 function. However, in many cases these reagents have not been rigorously validated, leading to potentially misleading results. In this report we tested several commercially-available antibodies for GCN5L1, and found that two-thirds of those available did not unambiguously detect the protein by western blot in cultured mouse cells or ex vivo liver tissue. These data suggest that previously published studies which used these unverified antibodies to measure GCN5L1 protein abundance, in the absence of other independent methods of corroboration, should be interpreted with appropriate caution.


Assuntos
Anticorpos , Animais , Camundongos , Anticorpos/imunologia , Anticorpos/metabolismo , Fígado/metabolismo , Fígado/imunologia , Camundongos Knockout , Proteínas Mitocondriais/imunologia , Proteínas do Tecido Nervoso/imunologia
19.
Sci Transl Med ; 16(741): eadj9052, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38569016

RESUMO

Microglia help limit the progression of Alzheimer's disease (AD) by constraining amyloid-ß (Aß) pathology, effected through a balance of activating and inhibitory intracellular signals delivered by distinct cell surface receptors. Human leukocyte Ig-like receptor B4 (LILRB4) is an inhibitory receptor of the immunoglobulin (Ig) superfamily that is expressed on myeloid cells and recognizes apolipoprotein E (ApoE) among other ligands. Here, we find that LILRB4 is highly expressed in the microglia of patients with AD. Using mice that accumulate Aß and carry a transgene encompassing a portion of the LILR region that includes LILRB4, we corroborated abundant LILRB4 expression in microglia wrapping around Aß plaques. Systemic treatment of these mice with an anti-human LILRB4 monoclonal antibody (mAb) reduced Aß load, mitigated some Aß-related behavioral abnormalities, enhanced microglia activity, and attenuated expression of interferon-induced genes. In vitro binding experiments established that human LILRB4 binds both human and mouse ApoE and that anti-human LILRB4 mAb blocks such interaction. In silico modeling, biochemical, and mutagenesis analyses identified a loop between the two extracellular Ig domains of LILRB4 required for interaction with mouse ApoE and further indicated that anti-LILRB4 mAb may block LILRB4-mApoE by directly binding this loop. Thus, targeting LILRB4 may be a potential therapeutic avenue for AD.


Assuntos
Doença de Alzheimer , Microglia , Humanos , Camundongos , Animais , Microglia/metabolismo , Anticorpos/metabolismo , Receptores de Superfície Celular/metabolismo , Amiloide/metabolismo , Modelos Animais de Doenças , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E , Leucócitos/metabolismo , Camundongos Transgênicos , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo
20.
Methods Mol Biol ; 2790: 405-416, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38649583

RESUMO

Antibodies are a valuable research tool, with uses including detection and quantification of specific proteins. By using peptide fragments to raise antibodies, they can be designed to differentiate between structurally similar proteins, or to bind conserved motifs in divergent proteins. Peptide sequence selection and antibody validation are crucial to ensure reliable results from antibody-based experiments. This chapter describes the steps for the identification of peptide sequences to produce protein- or isoform-specific antibodies using recombinant technologies as well as the subsequent validation of such antibodies. The photosynthetic protein Rubisco activase is used as a case study to explain the various steps involved and key aspects to take into consideration.


Assuntos
Anticorpos , Isoformas de Proteínas , Anticorpos/química , Anticorpos/imunologia , Anticorpos/metabolismo , Fotossíntese , Sequência de Aminoácidos , Proteínas de Plantas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...