Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.737
Filtrar
1.
BMC Genomics ; 25(1): 406, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724906

RESUMEN

Most proteins exert their functions by interacting with other proteins, making the identification of protein-protein interactions (PPI) crucial for understanding biological activities, pathological mechanisms, and clinical therapies. Developing effective and reliable computational methods for predicting PPI can significantly reduce the time-consuming and labor-intensive associated traditional biological experiments. However, accurately identifying the specific categories of protein-protein interactions and improving the prediction accuracy of the computational methods remain dual challenges. To tackle these challenges, we proposed a novel graph neural network method called GNNGL-PPI for multi-category prediction of PPI based on global graphs and local subgraphs. GNNGL-PPI consisted of two main components: using Graph Isomorphism Network (GIN) to extract global graph features from PPI network graph, and employing GIN As Kernel (GIN-AK) to extract local subgraph features from the subgraphs of protein vertices. Additionally, considering the imbalanced distribution of samples in each category within the benchmark datasets, we introduced an Asymmetric Loss (ASL) function to further enhance the predictive performance of the method. Through evaluations on six benchmark test sets formed by three different dataset partitioning algorithms (Random, BFS, DFS), GNNGL-PPI outperformed the state-of-the-art multi-category prediction methods of PPI, as measured by the comprehensive performance evaluation metric F1-measure. Furthermore, interpretability analysis confirmed the effectiveness of GNNGL-PPI as a reliable multi-category prediction method for predicting protein-protein interactions.


Asunto(s)
Algoritmos , Biología Computacional , Redes Neurales de la Computación , Mapeo de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Biología Computacional/métodos , Mapas de Interacción de Proteínas , Humanos , Proteínas/metabolismo
2.
Methods Mol Biol ; 2807: 245-258, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743233

RESUMEN

The study of host-pathogen interaction often requires interrogating the protein-protein interactions and examining post-translational modifications of the proteins. Traditional protein detection strategies are limited in their sensitivity, specificity, and multiplexing capabilities. The Proximity Ligation Assay (PLA), a versatile and powerful molecular technique, can overcome these limitations. PLA blends the specificity of antibodies, two antibodies detecting two different epitopes on the same or two different proteins, with the amplification efficiency of a polymerase to allow highly specific and sensitive detection of low-abundant proteins, protein-protein interactions, or protein modifications. In this protocol, we describe the application of PLA to detect the proximity between HIV-1 Tat with one of its cellular partners, p65, in an infected host cell. The protocol could be applied to any other context with slight modifications. Of note, PLA can only confirm the physical proximity between two epitopes or proteins; however, the proximity need not necessarily allude to the functional interaction between the two proteins.


Asunto(s)
VIH-1 , Interacciones Huésped-Patógeno , Humanos , VIH-1/inmunología , Mapeo de Interacción de Proteínas/métodos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Infecciones por VIH/virología , Unión Proteica
3.
Methods Mol Biol ; 2808: 9-17, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743359

RESUMEN

Protein-fragment complementation assays (PCAs) are powerful tools to investigate protein-protein interactions in a cellular context. These are especially useful to study unstable proteins and weak interactions that may not resist protein isolation or purification. The PCA based on the reconstitution of the Gaussia princeps luciferase (split-luc) is a sensitive approach allowing the mapping of protein-protein interactions and the semiquantitative measurement of binding affinity. Here, we describe the split-luc protocol we used to map the viral interactome of measles virus polymerase complex.


Asunto(s)
Virus del Sarampión , Unión Proteica , Mapeo de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Humanos , Luciferasas/metabolismo , Luciferasas/genética , Proteínas Virales/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo
4.
Methods Mol Biol ; 2808: 89-103, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743364

RESUMEN

The study of virus-host interactions is essential to achieve a comprehensive understanding of the viral replication process. The commonly used methods are yeast two-hybrid approach and transient expression of a single tagged viral protein in host cells followed by affinity purification of interacting cellular proteins and mass spectrometry analysis (AP-MS). However, by these approaches, virus-host protein-protein interactions are detected in the absence of a real infection, not always correctly compartmentalized, and for the yeast two-hybrid approach performed in a heterologous system. Thus, some of the detected protein-protein interactions may be artificial. Here we describe a new strategy based on recombinant viruses expressing tagged viral proteins to capture both direct and indirect protein partners during the infection (AP-MS in viral context). This way, virus-host protein-protein interacting co-complexes can be purified directly from infected cells for further characterization.


Asunto(s)
Interacciones Huésped-Patógeno , Virus del Sarampión , Genética Inversa , Proteínas Virales , Virus del Sarampión/genética , Humanos , Interacciones Huésped-Patógeno/genética , Genética Inversa/métodos , Proteínas Virales/metabolismo , Proteínas Virales/genética , Técnicas del Sistema de Dos Híbridos , Replicación Viral , Espectrometría de Masas , Mapeo de Interacción de Proteínas/métodos , Sarampión/virología , Sarampión/metabolismo , Animales , Unión Proteica
5.
Proc Natl Acad Sci U S A ; 121(21): e2400260121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38743624

RESUMEN

We introduce ZEPPI (Z-score Evaluation of Protein-Protein Interfaces), a framework to evaluate structural models of a complex based on sequence coevolution and conservation involving residues in protein-protein interfaces. The ZEPPI score is calculated by comparing metrics for an interface to those obtained from randomly chosen residues. Since contacting residues are defined by the structural model, this obviates the need to account for indirect interactions. Further, although ZEPPI relies on species-paired multiple sequence alignments, its focus on interfacial residues allows it to leverage quite shallow alignments. ZEPPI can be implemented on a proteome-wide scale and is applied here to millions of structural models of dimeric complexes in the Escherichia coli and human interactomes found in the PrePPI database. PrePPI's scoring function is based primarily on the evaluation of protein-protein interfaces, and ZEPPI adds a new feature to this analysis through the incorporation of evolutionary information. ZEPPI performance is evaluated through applications to experimentally determined complexes and to decoys from the CASP-CAPRI experiment. As we discuss, the standard CAPRI scores used to evaluate docking models are based on model quality and not on the ability to give yes/no answers as to whether two proteins interact. ZEPPI is able to detect weak signals from PPI models that the CAPRI scores define as incorrect and, similarly, to identify potential PPIs defined as low confidence by the current PrePPI scoring function. A number of examples that illustrate how the combination of PrePPI and ZEPPI can yield functional hypotheses are provided.


Asunto(s)
Proteoma , Proteoma/metabolismo , Humanos , Mapeo de Interacción de Proteínas/métodos , Modelos Moleculares , Escherichia coli/metabolismo , Escherichia coli/genética , Bases de Datos de Proteínas , Unión Proteica , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas/química , Proteínas/metabolismo , Alineación de Secuencia
6.
BMC Genomics ; 25(1): 466, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741045

RESUMEN

BACKGROUND: Protein-protein interactions (PPIs) hold significant importance in biology, with precise PPI prediction as a pivotal factor in comprehending cellular processes and facilitating drug design. However, experimental determination of PPIs is laborious, time-consuming, and often constrained by technical limitations. METHODS: We introduce a new node representation method based on initial information fusion, called FFANE, which amalgamates PPI networks and protein sequence data to enhance the precision of PPIs' prediction. A Gaussian kernel similarity matrix is initially established by leveraging protein structural resemblances. Concurrently, protein sequence similarities are gauged using the Levenshtein distance, enabling the capture of diverse protein attributes. Subsequently, to construct an initial information matrix, these two feature matrices are merged by employing weighted fusion to achieve an organic amalgamation of structural and sequence details. To gain a more profound understanding of the amalgamated features, a Stacked Autoencoder (SAE) is employed for encoding learning, thereby yielding more representative feature representations. Ultimately, classification models are trained to predict PPIs by using the well-learned fusion feature. RESULTS: When employing 5-fold cross-validation experiments on SVM, our proposed method achieved average accuracies of 94.28%, 97.69%, and 84.05% in terms of Saccharomyces cerevisiae, Homo sapiens, and Helicobacter pylori datasets, respectively. CONCLUSION: Experimental findings across various authentic datasets validate the efficacy and superiority of this fusion feature representation approach, underscoring its potential value in bioinformatics.


Asunto(s)
Biología Computacional , Mapeo de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Biología Computacional/métodos , Algoritmos , Helicobacter pylori/metabolismo , Helicobacter pylori/genética , Máquina de Vectores de Soporte , Proteínas/metabolismo , Proteínas/química , Humanos , Mapas de Interacción de Proteínas , Bases de Datos de Proteínas
7.
Methods Mol Biol ; 2787: 305-313, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38656499

RESUMEN

Bimolecular fluorescence complementation (BiFC) is a powerful tool for studying protein-protein interactions in living cells. By fusing interacting proteins to fluorescent protein fragments, BiFC allows visualization of spatial localization patterns of protein complexes. This method has been adapted to a variety of expression systems in different organisms and is widely used to study protein interactions in plant cells. The Agrobacterium-mediated transient expression protocol for BiFC assays in Nicotiana benthamiana (N. benthamiana) leaf cells is widely used, but in this chapter, a method for BiFC assay using Arabidopsis thaliana protoplasts is presented.


Asunto(s)
Arabidopsis , Hojas de la Planta , Protoplastos , Arabidopsis/metabolismo , Arabidopsis/genética , Protoplastos/metabolismo , Hojas de la Planta/metabolismo , Hojas de la Planta/genética , Mapeo de Interacción de Proteínas/métodos , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Microscopía Fluorescente/métodos , Proteínas Luminiscentes/metabolismo , Proteínas Luminiscentes/genética , Nicotiana/metabolismo , Nicotiana/genética , Unión Proteica , Agrobacterium/genética , Agrobacterium/metabolismo
8.
J Am Soc Mass Spectrom ; 35(5): 1055-1058, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38606722

RESUMEN

Proximity labeling techniques, such as APEX-MS, provide valuable insights into proximal interactome mapping; however, the verification of biotinylated peptides is not straightforward. With this as motivation, we present a new module integrated into PatternLab for proteomics to enable APEX-MS data interpretation by targeting diagnostic fragment ions associated with APEX modifications. We reanalyzed a previously published APEX-MS data set and report a significant number of biotinylated peptides and, consequently, a confident set of proximal proteins. As the module is part of the widely adopted PatternLab for proteomics software suite, it offers users a comprehensive, easy, and integrated solution for data analysis. Given the broad utility of the APEX-MS technique in various biological contexts, we anticipate that our module will be a valuable asset to researchers, facilitating and enhancing interactome studies. PatternLab's APEX, including a usage protocol, is available at http://patternlabforproteomics.org/apex.


Asunto(s)
Proteómica , Programas Informáticos , Proteómica/métodos , Espectrometría de Masas/métodos , Humanos , Mapeo de Interacción de Proteínas/métodos , Biotinilación , Péptidos/análisis , Péptidos/química , Péptidos/metabolismo
9.
BMC Bioinformatics ; 25(1): 157, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38643108

RESUMEN

BACKGROUND: The identification of essential proteins can help in understanding the minimum requirements for cell survival and development to discover drug targets and prevent disease. Nowadays, node ranking methods are a common way to identify essential proteins, but the poor data quality of the underlying PIN has somewhat hindered the identification accuracy of essential proteins for these methods in the PIN. Therefore, researchers constructed refinement networks by considering certain biological properties of interacting protein pairs to improve the performance of node ranking methods in the PIN. Studies show that proteins in a complex are more likely to be essential than proteins not present in the complex. However, the modularity is usually ignored for the refinement methods of the PINs. METHODS: Based on this, we proposed a network refinement method based on module discovery and biological information. The idea is, first, to extract the maximal connected subgraph in the PIN, and to divide it into different modules by using Fast-unfolding algorithm; then, to detect critical modules according to the orthologous information, subcellular localization information and topology information within each module; finally, to construct a more refined network (CM-PIN) by using the identified critical modules. RESULTS: To evaluate the effectiveness of the proposed method, we used 12 typical node ranking methods (LAC, DC, DMNC, NC, TP, LID, CC, BC, PR, LR, PeC, WDC) to compare the overall performance of the CM-PIN with those on the S-PIN, D-PIN and RD-PIN. The experimental results showed that the CM-PIN was optimal in terms of the identification number of essential proteins, precision-recall curve, Jackknifing method and other criteria, and can help to identify essential proteins more accurately.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Mapeo de Interacción de Proteínas/métodos , Algoritmos , Mapas de Interacción de Proteínas , Biología Computacional/métodos
10.
Methods Mol Biol ; 2806: 219-227, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38676806

RESUMEN

Proteins are large, complex molecules that regulate multiple functions within the cell. The protein rarely functions as a single molecule, but rather interacts with one or more other proteins forming a dynamic network. Protein-protein interactions are critical for regulating the cell's response toward various stimuli from outside and inside the cell. Identification of protein-protein interactions enhanced our understanding of various biological processes in the living cell. Immunoprecipitation (IP) has been one of the standard and most commonly used biochemical methods to identify and confirm protein-protein interactions. IP uses a target protein-specific antibody conjugated with protein A/G affinity beads to identify molecules interacting with the target protein. Here, we describe the principle, procedure and challenges of the IP assay.


Asunto(s)
Inmunoprecipitación , Mapeo de Interacción de Proteínas , Inmunoprecipitación/métodos , Humanos , Animales , Mapeo de Interacción de Proteínas/métodos , Ratones , Unión Proteica , Xenoinjertos , Proteínas/metabolismo
11.
Nat Commun ; 15(1): 3516, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664367

RESUMEN

Chemical cross-linking reactions (XL) are an important strategy for studying protein-protein interactions (PPIs), including low abundant sub-complexes, in structural biology. However, choosing XL reagents and conditions is laborious and mostly limited to analysis of protein assemblies that can be resolved using SDS-PAGE. To overcome these limitations, we develop here a denaturing mass photometry (dMP) method for fast, reliable and user-friendly optimization and monitoring of chemical XL reactions. The dMP is a robust 2-step protocol that ensures 95% of irreversible denaturation within only 5 min. We show that dMP provides accurate mass identification across a broad mass range (30 kDa-5 MDa) along with direct label-free relative quantification of all coexisting XL species (sub-complexes and aggregates). We compare dMP with SDS-PAGE and observe that, unlike the benchmark, dMP is time-efficient (3 min/triplicate), requires significantly less material (20-100×) and affords single molecule sensitivity. To illustrate its utility for routine structural biology applications, we show that dMP affords screening of 20 XL conditions in 1 h, accurately identifying and quantifying all coexisting species. Taken together, we anticipate that dMP will have an impact on ability to structurally characterize more PPIs and macromolecular assemblies, expected final complexes but also sub-complexes that form en route.


Asunto(s)
Reactivos de Enlaces Cruzados , Fotometría , Desnaturalización Proteica , Reactivos de Enlaces Cruzados/química , Fotometría/métodos , Proteínas/química , Proteínas/metabolismo , Electroforesis en Gel de Poliacrilamida/métodos , Mapeo de Interacción de Proteínas/métodos , Espectrometría de Masas/métodos , Humanos
12.
Nat Commun ; 15(1): 2875, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570497

RESUMEN

The characterization of protein-protein interactions (PPIs) is fundamental to the understanding of biochemical processes. Many methods have been established to identify and study direct PPIs; however, screening and investigating PPIs involving large or poorly soluble proteins remains challenging. Here, we introduce ReLo, a simple, rapid, and versatile cell culture-based method for detecting and investigating interactions in a cellular context. Our experiments demonstrate that ReLo specifically detects direct binary PPIs. Furthermore, we show that ReLo bridging experiments can also be used to determine the binding topology of subunits within multiprotein complexes. In addition, ReLo facilitates the identification of protein domains that mediate complex formation, allows screening for interfering point mutations, and it is sensitive to drugs that mediate or disrupt an interaction. In summary, ReLo is a simple and rapid alternative for the study of PPIs, especially when studying structurally complex proteins or when established methods fail.


Asunto(s)
Mapeo de Interacción de Proteínas , Proteínas , Mapeo de Interacción de Proteínas/métodos , Proteínas/metabolismo
13.
BMC Bioinformatics ; 25(1): 172, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689238

RESUMEN

BACKGROUND: Protein-protein interactions (PPIs) are conveyed through binding interfaces or surface patches on proteins that become buried upon binding. Structural and biophysical analysis of many protein-protein interfaces revealed certain unique features of these surfaces that determine the energetics of interactions and play a critical role in protein evolution. One of the significant aspects of binding interfaces is the presence of binding hot spots, where mutations are highly deleterious for binding. Conversely, binding cold spots are positions occupied by suboptimal amino acids and several mutations in such positions could lead to affinity enhancement. While there are many software programs for identification of hot spot positions, there is currently a lack of software for cold spot detection. RESULTS: In this paper, we present Cold Spot SCANNER, a Colab Notebook, which scans a PPI binding interface and identifies cold spots resulting from cavities, unfavorable charge-charge, and unfavorable charge-hydrophobic interactions. The software offers a Py3DMOL-based interface that allows users to visualize cold spots in the context of the protein structure and generates a zip file containing the results for easy download. CONCLUSIONS: Cold spot identification is of great importance to protein engineering studies and provides a useful insight into protein evolution. Cold Spot SCANNER is open to all users without login requirements and can be accessible at: https://colab. RESEARCH: google.com/github/sagagugit/Cold-Spot-Scanner/blob/main/Cold_Spot_Scanner.ipynb .


Asunto(s)
Proteínas , Programas Informáticos , Proteínas/química , Proteínas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Unión Proteica , Conformación Proteica , Modelos Moleculares , Sitios de Unión , Interacciones Hidrofóbicas e Hidrofílicas
14.
Drug Discov Today ; 29(5): 103979, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38608830

RESUMEN

Drug discovery often begins with a new target. Protein-protein interactions (PPIs) are crucial to multitudinous cellular processes and offer a promising avenue for drug-target discovery. PPIs are characterized by multi-level complexity: at the protein level, interaction networks can be used to identify potential targets, whereas at the residue level, the details of the interactions of individual PPIs can be used to examine a target's druggability. Much great progress has been made in target discovery through multi-level PPI-related computational approaches, but these resources have not been fully discussed. Here, we systematically survey bioinformatics tools for identifying and assessing potential drug targets, examining their characteristics, limitations and applications. This work will aid the integration of the broader protein-to-network context with the analysis of detailed binding mechanisms to support the discovery of drug targets.


Asunto(s)
Biología Computacional , Descubrimiento de Drogas , Descubrimiento de Drogas/métodos , Biología Computacional/métodos , Humanos , Proteínas/metabolismo , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapeo de Interacción de Proteínas/métodos , Unión Proteica
15.
Comput Methods Programs Biomed ; 250: 108188, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38657382

RESUMEN

BACKGROUND AND OBJECTIVE: The protein-protein interaction (PPI) network alignment has proven to be an efficient technique in the diagnosis and prevention of certain diseases. However, the difficulty in maximizing, at the same time, the two qualities that measure the goodness of alignments (topological and biological quality) has led aligners to produce very different alignments. Thus making a comparative study among alignments of such different qualities a big challenge. Multi-objective optimization is a computer method, which is very powerful in this kind of contexts because both conflicting qualities are considered together. Analysing the alignments of each PPI network aligner with multi-objective methodologies allows you to visualize a bigger picture of the alignments and their qualities, obtaining very interesting conclusions. This paper proposes a comprehensive PPI network aligner study in the multi-objective domain. METHODS: Alignments from each aligner and all aligners together were studied and compared to each other via Pareto dominance methodologies. The best alignments produced by each aligner and all aligners together for five different alignment scenarios were displayed in Pareto front graphs. Later, the aligners were ranked according to the topological, biological, and combined quality of their alignments. Finally, the aligners were also ranked based on their average runtimes. RESULTS: Regarding aligners constructing the best overall alignments, we found that SAlign, BEAMS, SANA, and HubAlign are the best options. Additionally, the alignments of best topological quality are produced by: SANA, SAlign, and HubAlign aligners. On the contrary, the aligners returning the alignments of best biological quality are: BEAMS, TAME, and WAVE. However, if there are time constraints, it is recommended to select SAlign to obtain high topological quality alignments and PISwap or SAlign aligners for high biological quality alignments. CONCLUSIONS: The use of the SANA aligner is recommended for obtaining the best alignments of topological quality, BEAMS for alignments of the best biological quality, and SAlign for alignments of the best combined topological and biological quality. Simultaneously, SANA and BEAMS have above-average runtimes. Therefore, it is suggested, if necessary due to time restrictions, to choose other, faster aligners like SAlign or PISwap whose alignments are also of high quality.


Asunto(s)
Algoritmos , Mapeo de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , Programas Informáticos , Alineación de Secuencia , Biología Computacional/métodos , Proteínas/química , Proteínas/metabolismo , Humanos
16.
J Chem Inf Model ; 64(8): 3332-3349, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38470439

RESUMEN

Analyzing the similarity of protein interfaces in protein-protein interactions gives new insights into protein function and assists in discovering new drugs. Usually, tools that assess the similarity focus on the interactions between two protein interfaces, while sometimes we only have one predicted interface. Herein, we present PiMine, a database-driven protein interface similarity search. It compares interface residues of one or two interacting chains by calculating and searching tetrahedral geometric patterns of α-carbon atoms and calculating physicochemical and shape-based similarity. On a dedicated, tailor-made dataset, we show that PiMine outperforms commonly used comparison tools in terms of early enrichment when considering interfaces of sequentially and structurally unrelated proteins. In an application example, we demonstrate its usability for protein interaction partner prediction by comparing predicted interfaces to known protein-protein interfaces.


Asunto(s)
Bases de Datos de Proteínas , Proteínas , Proteínas/química , Proteínas/metabolismo , Conformación Proteica , Unión Proteica , Mapeo de Interacción de Proteínas/métodos , Modelos Moleculares
17.
Nucleic Acids Res ; 52(8): e42, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38512053

RESUMEN

We present txtools, an R package that enables the processing, analysis, and visualization of RNA-seq data at the nucleotide-level resolution, seamlessly integrating alignments to the genome with transcriptomic representation. txtools' main inputs are BAM files and a transcriptome annotation, and the main output is a table, capturing mismatches, deletions, and the number of reads beginning and ending at each nucleotide in the transcriptomic space. txtools further facilitates downstream visualization and analyses. We showcase, using examples from the epitranscriptomic field, how a few calls to txtools functions can yield insightful and ready-to-publish results. txtools is of broad utility also in the context of structural mapping and RNA:protein interaction mapping. By providing a simple and intuitive framework, we believe that txtools will be a useful and convenient tool and pave the path for future discovery. txtools is available for installation from its GitHub repository at https://github.com/AngelCampos/txtools.


Asunto(s)
ARN , Programas Informáticos , ARN/química , ARN/genética , ARN/metabolismo , Humanos , Transcriptoma , Procesamiento Postranscripcional del ARN , Análisis de Secuencia de ARN/métodos , RNA-Seq/métodos , Conformación de Ácido Nucleico , Mapeo de Interacción de Proteínas/métodos
18.
Comput Biol Med ; 172: 108287, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38503089

RESUMEN

Protein-protein interactions (PPIs) have shown increasing potential as novel drug targets. The design and development of small molecule inhibitors targeting specific PPIs are crucial for the prevention and treatment of related diseases. Accordingly, effective computational methods are highly desired to meet the emerging need for the large-scale accurate prediction of PPI inhibitors. However, existing machine learning models rely heavily on the manual screening of features and lack generalizability. Here, we propose a new PPI inhibitor prediction method based on autoencoders with adversarial training (named PPII-AEAT) that can adaptively learn molecule representation to cope with different PPI targets. First, Extended-connectivity fingerprints and Mordred descriptors are employed to extract the primary features of small molecular compounds. Then, an autoencoder architecture is trained in three phases to learn high-level representations and predict inhibitory scores. We evaluate PPII-AEAT on nine PPI targets and two different tasks, including the PPI inhibitor identification task and inhibitory potency prediction task. The experimental results show that our proposed PPII-AEAT outperforms state-of-the-art methods.


Asunto(s)
Aprendizaje Automático , Mapeo de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos
19.
J Chem Inf Model ; 64(8): 2979-2987, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38526504

RESUMEN

Proteins are vital components of the biological world and serve a multitude of functions. They interact with other molecules through their interfaces and participate in crucial cellular processes. Disruption of these interactions can have negative effects on organisms, highlighting the importance of studying protein-protein interfaces for developing targeted therapies for diseases. Therefore, the development of a reliable method for investigating protein-protein interactions is of paramount importance. In this work, we present an approach for validating protein-protein interfaces using learned interface representations. The approach involves using a graph-based contrastive autoencoder architecture and a transformer to learn representations of protein-protein interaction interfaces from unlabeled data and then validating them through learned representations with a graph neural network. Our method achieves an accuracy of 0.91 for the test set, outperforming existing GNN-based methods. We demonstrate the effectiveness of our approach on a benchmark data set and show that it provides a promising solution for validating protein-protein interfaces.


Asunto(s)
Mapeo de Interacción de Proteínas , Proteínas , Proteínas/química , Proteínas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Redes Neurales de la Computación , Unión Proteica , Bases de Datos de Proteínas , Modelos Moleculares
20.
Bioinformatics ; 40(3)2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38449296

RESUMEN

MOTIVATION: The functional complexity of biochemical processes is strongly related to the interplay of proteins and their assembly into protein complexes. In recent years, the discovery and characterization of protein complexes have substantially progressed through advances in cryo-electron microscopy, proteomics, and computational structure prediction. This development results in a strong need for computational approaches to analyse the data of large protein complexes for structural and functional characterization. Here, we aim to provide a suitable approach, which processes the growing number of large protein complexes, to obtain biologically meaningful information on the hierarchical organization of the structures of protein complexes. RESULTS: We modelled the quaternary structure of protein complexes as undirected, labelled graphs called complex graphs. In complex graphs, the vertices represent protein chains and the edges spatial chain-chain contacts. We hypothesized that clusters based on the complex graph correspond to functional biological modules. To compute the clusters, we applied the Leiden clustering algorithm. To evaluate our approach, we chose the human respiratory complex I, which has been extensively investigated and exhibits a known biological module structure experimentally validated. Additionally, we characterized a eukaryotic group II chaperonin TRiC/CCT and the head of the bacteriophage Φ29. The analysis of the protein complexes correlated with experimental findings and indicated known functional, biological modules. Using our approach enables not only to predict functional biological modules in large protein complexes with characteristic features but also to investigate the flexibility of specific regions and coformational changes. The predicted modules can aid in the planning and analysis of experiments. AVAILABILITY AND IMPLEMENTATION: Jupyter notebooks to reproduce the examples are available on our public GitHub repository: https://github.com/MolBIFFM/PTGLtools/tree/main/PTGLmodulePrediction.


Asunto(s)
Biología Computacional , Mapeo de Interacción de Proteínas , Humanos , Mapeo de Interacción de Proteínas/métodos , Microscopía por Crioelectrón , Biología Computacional/métodos , Algoritmos , Proteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...