Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 5.746
1.
Comput Biol Med ; 177: 108669, 2024 Jul.
Article En | MEDLINE | ID: mdl-38833802

The process of experimentally confirming complex interaction networks among proteins is time-consuming and laborious. This study aims to address Protein-Protein Interactions (PPIs) prediction based on graph neural networks (GNN). A novel multilevel prediction model for PPIs named DSSGNN-PPI (Double Structure and Sequence GNN for PPIs) is designed. Initially, a distance graph between amino acid residues is constructed. Subsequently, the distance graph is fed into an underlying graph attention network module. This enables us to efficiently learn vector representations that encode the three-dimensional structure of proteins and simultaneously aggregate key local patterns and overall topological information to obtain graph embedding that adequately represent local and global structural features. In addition, the embedding representations that reflect sequence properties are obtained. Two features are fused to construct high-level protein complex networks, which are fed into the designed gated graph attention network to extract complex topological patterns. By combining heterogeneous multi-source information from downstream structure graph and upstream sequence models, the understanding of PPIs is comprehensively enhanced. A series of evaluation results validate the remarkable effectiveness of DSSGNN-PPI framework in enhancing the prediction of multi-type interactions among proteins. The multilevel representation learning and information fusion strategies provide a new effective solution paradigm for structural biology problems. The source code for DSSGNN-PPI has been hosted on GitHub and is available at https://github.com/cstudy1/DSSGNN-PPI.


Neural Networks, Computer , Protein Interaction Mapping , Proteins , Protein Interaction Mapping/methods , Proteins/chemistry , Proteins/metabolism , Protein Interaction Maps , Computational Biology/methods , Humans , Databases, Protein
2.
Brief Bioinform ; 25(4)2024 May 23.
Article En | MEDLINE | ID: mdl-38856171

The identification of protein complexes from protein interaction networks is crucial in the understanding of protein function, cellular processes and disease mechanisms. Existing methods commonly rely on the assumption that protein interaction networks are highly reliable, yet in reality, there is considerable noise in the data. In addition, these methods fail to account for the regulatory roles of biomolecules during the formation of protein complexes, which is crucial for understanding the generation of protein interactions. To this end, we propose a SpatioTemporal constrained RNA-protein heterogeneous network for Protein Complex Identification (STRPCI). STRPCI first constructs a multiplex heterogeneous protein information network to capture deep semantic information by extracting spatiotemporal interaction patterns. Then, it utilizes a dual-view aggregator to aggregate heterogeneous neighbor information from different layers. Finally, through contrastive learning, STRPCI collaboratively optimizes the protein embedding representations under different spatiotemporal interaction patterns. Based on the protein embedding similarity, STRPCI reweights the protein interaction network and identifies protein complexes with core-attachment strategy. By considering the spatiotemporal constraints and biomolecular regulatory factors of protein interactions, STRPCI measures the tightness of interactions, thus mitigating the impact of noisy data on complex identification. Evaluation results on four real PPI networks demonstrate the effectiveness and strong biological significance of STRPCI. The source code implementation of STRPCI is available from https://github.com/LI-jasm/STRPCI.


Protein Interaction Maps , RNA , RNA/metabolism , RNA/chemistry , Proteins/metabolism , Proteins/chemistry , Computational Biology/methods , Algorithms , Protein Interaction Mapping/methods , Humans
3.
Brief Bioinform ; 25(4)2024 May 23.
Article En | MEDLINE | ID: mdl-38851299

Protein-protein interactions (PPIs) are the basis of many important biological processes, with protein complexes being the key forms implementing these interactions. Understanding protein complexes and their functions is critical for elucidating mechanisms of life processes, disease diagnosis and treatment and drug development. However, experimental methods for identifying protein complexes have many limitations. Therefore, it is necessary to use computational methods to predict protein complexes. Protein sequences can indicate the structure and biological functions of proteins, while also determining their binding abilities with other proteins, influencing the formation of protein complexes. Integrating these characteristics to predict protein complexes is very promising, but currently there is no effective framework that can utilize both protein sequence and PPI network topology for complex prediction. To address this challenge, we have developed HyperGraphComplex, a method based on hypergraph variational autoencoder that can capture expressive features from protein sequences without feature engineering, while also considering topological properties in PPI networks, to predict protein complexes. Experiment results demonstrated that HyperGraphComplex achieves satisfactory predictive performance when compared with state-of-art methods. Further bioinformatics analysis shows that the predicted protein complexes have similar attributes to known ones. Moreover, case studies corroborated the remarkable predictive capability of our model in identifying protein complexes, including 3 that were not only experimentally validated by recent studies but also exhibited high-confidence structural predictions from AlphaFold-Multimer. We believe that the HyperGraphComplex algorithm and our provided proteome-wide high-confidence protein complex prediction dataset will help elucidate how proteins regulate cellular processes in the form of complexes, and facilitate disease diagnosis and treatment and drug development. Source codes are available at https://github.com/LiDlab/HyperGraphComplex.


Computational Biology , Protein Interaction Mapping , Computational Biology/methods , Protein Interaction Mapping/methods , Proteins/metabolism , Proteins/chemistry , Algorithms , Protein Interaction Maps , Databases, Protein , Humans , Sequence Analysis, Protein/methods , Amino Acid Sequence
4.
BMC Genomics ; 25(1): 466, 2024 May 13.
Article En | MEDLINE | ID: mdl-38741045

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.


Computational Biology , Protein Interaction Mapping , Protein Interaction Mapping/methods , Computational Biology/methods , Algorithms , Helicobacter pylori/metabolism , Helicobacter pylori/genetics , Support Vector Machine , Proteins/metabolism , Proteins/chemistry , Humans , Protein Interaction Maps , Databases, Protein
5.
Proc Natl Acad Sci U S A ; 121(21): e2400260121, 2024 May 21.
Article En | MEDLINE | ID: mdl-38743624

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.


Proteome , Proteome/metabolism , Humans , Protein Interaction Mapping/methods , Models, Molecular , Escherichia coli/metabolism , Escherichia coli/genetics , Databases, Protein , Protein Binding , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Proteins/chemistry , Proteins/metabolism , Sequence Alignment
6.
BMC Genomics ; 25(1): 406, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724906

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.


Algorithms , Computational Biology , Neural Networks, Computer , Protein Interaction Mapping , Protein Interaction Mapping/methods , Computational Biology/methods , Protein Interaction Maps , Humans , Proteins/metabolism
7.
Methods Mol Biol ; 2807: 245-258, 2024.
Article En | MEDLINE | ID: mdl-38743233

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.


HIV-1 , Host-Pathogen Interactions , Humans , HIV-1/immunology , Protein Interaction Mapping/methods , tat Gene Products, Human Immunodeficiency Virus/metabolism , HIV Infections/virology , Protein Binding
8.
Methods Mol Biol ; 2808: 9-17, 2024.
Article En | MEDLINE | ID: mdl-38743359

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.


Measles virus , Protein Binding , Protein Interaction Mapping , Protein Interaction Mapping/methods , Humans , Luciferases/metabolism , Luciferases/genetics , Viral Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism
9.
Methods Mol Biol ; 2808: 89-103, 2024.
Article En | MEDLINE | ID: mdl-38743364

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.


Host-Pathogen Interactions , Measles virus , Reverse Genetics , Viral Proteins , Measles virus/genetics , Humans , Host-Pathogen Interactions/genetics , Reverse Genetics/methods , Viral Proteins/metabolism , Viral Proteins/genetics , Two-Hybrid System Techniques , Virus Replication , Mass Spectrometry , Protein Interaction Mapping/methods , Measles/virology , Measles/metabolism , Animals , Protein Binding
10.
J Theor Biol ; 589: 111850, 2024 Jul 21.
Article En | MEDLINE | ID: mdl-38740126

Protein-protein interactions (PPIs) are crucial for various biological processes, and predicting PPIs is a major challenge. To solve this issue, the most common method is link prediction. Currently, the link prediction methods based on network Paths of Length Three (L3) have been proven to be highly effective. In this paper, we propose a novel link prediction algorithm, named SMS, which is based on L3 and protein similarities. We first design a mixed similarity that combines the topological structure and attribute features of nodes. Then, we compute the predicted value by summing the product of all similarities along the L3. Furthermore, we propose the Max Similarity Multiplied Similarity (maxSMS) algorithm from the perspective of maximum impact. Our computational prediction results show that on six datasets, including S. cerevisiae, H. sapiens, and others, the maxSMS algorithm improves the precision of the top 500, area under the precision-recall curve, and normalized discounted cumulative gain by an average of 26.99%, 53.67%, and 6.7%, respectively, compared to other optimal methods.


Algorithms , Protein Interaction Mapping , Protein Interaction Maps , Humans , Protein Interaction Mapping/methods , Computational Biology/methods , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Databases, Protein , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
11.
Comput Biol Med ; 176: 108543, 2024 Jun.
Article En | MEDLINE | ID: mdl-38744015

Proteins play a vital role in various biological processes and achieve their functions through protein-protein interactions (PPIs). Thus, accurate identification of PPI sites is essential. Traditional biological methods for identifying PPIs are costly, labor-intensive, and time-consuming. The development of computational prediction methods for PPI sites offers promising alternatives. Most known deep learning (DL) methods employ layer-wise multi-scale CNNs to extract features from protein sequences. But, these methods usually neglect the spatial positions and hierarchical information embedded within protein sequences, which are actually crucial for PPI site prediction. In this paper, we propose MR2CPPIS, a novel sequence-based DL model that utilizes the multi-scale Res2Net with coordinate attention mechanism to exploit multi-scale features and enhance PPI site prediction capability. We leverage the multi-scale Res2Net to expand the receptive field for each network layer, thus capturing multi-scale information of protein sequences at a granular level. To further explore the local contextual features of each target residue, we employ a coordinate attention block to characterize the precise spatial position information, enabling the network to effectively extract long-range dependencies. We evaluate our MR2CPPIS on three public benchmark datasets (Dset 72, Dset 186, and PDBset 164), achieving state-of-the-art performance. The source codes are available at https://github.com/YyinGong/MR2CPPIS.


Deep Learning , Proteins/metabolism , Proteins/chemistry , Protein Interaction Mapping/methods , Computational Biology/methods , Humans , Databases, Protein
12.
Comput Biol Med ; 177: 108623, 2024 Jul.
Article En | MEDLINE | ID: mdl-38788374

Prediction of protein-protein interaction (PPI) types enhances the comprehension of the underlying structural characteristics and functions of proteins, which gives rise to a multi-label classification problem. The nominal features describe the physicochemical characteristics of proteins directly, establishing a more robust correlation with the interaction types between proteins than ordered features. Motivated by this, we propose a multi-label PPI prediction model referred to as CoMPPI (Co-training based Multi-Label prediction of Protein-Protein Interaction). This approach aims to maximize the utility of both ordered and nominal features extracted from protein sequences. Specifically, CoMPPI incorporates graph convolutional network (GCN) and 1D convolution operation to process the complementary subsets of features individually, leveraging both local and contextualized information in a more efficient way. In addition, two multi-type PPI datasets were constructed to eliminate the duplication in previous datasets. We compare the performance of CoMPPI with three state-of-the-art methods on three datasets partitioned using distinct schemes (Breadth-first search, Depth-first search, and Random), CoMPPI consistently outperforms the other methods across all cases, demonstrating improvements ranging from 3.81% to 32.40% in Micro-F1. The subsequent ablation experiment confirms the efficacy of employing the co-training framework for multi-label PPI prediction, indicating promising avenues for future advancements in this domain.


Protein Interaction Mapping , Proteins , Proteins/chemistry , Proteins/metabolism , Protein Interaction Mapping/methods , Databases, Protein , Humans , Computational Biology/methods
13.
Curr Opin Struct Biol ; 86: 102822, 2024 06.
Article En | MEDLINE | ID: mdl-38685162

Protein-protein interactions (PPIs) play a critical role in cellular signaling and represent interesting targets for therapeutic intervention. 14-3-3 proteins integrate many signaling targets via PPIs and are frequently implicated in disease, making them intriguing drug targets. Here, we review the recent advances in the 14-3-3 field. It will discuss the roles 14-3-3 proteins play within the cell, elucidation of their expansive interactome, and the complex mechanisms that underpin their function. In addition, the review will discuss significant advances in the development of molecular glues that target 14-3-3 PPIs. In particular, it will focus on novel drug discovery and development methodologies that have delivered selective, potent, and drug-like molecules that could open new avenues for the development of precision molecular tools and medicines.


14-3-3 Proteins , Protein Interaction Maps , 14-3-3 Proteins/metabolism , Humans , Protein Binding , Drug Discovery , Signal Transduction , Animals , Protein Interaction Mapping/methods
14.
Bioinformatics ; 40(5)2024 Jan 05.
Article En | MEDLINE | ID: mdl-38640481

MOTIVATION: Protein-protein interaction sites (PPIS) are crucial for deciphering protein action mechanisms and related medical research, which is the key issue in protein action research. Recent studies have shown that graph neural networks have achieved outstanding performance in predicting PPIS. However, these studies often neglect the modeling of information at different scales in the graph and the symmetry of protein molecules within three-dimensional space. RESULTS: In response to this gap, this article proposes the MEG-PPIS approach, a PPIS prediction method based on multi-scale graph information and E(n) equivariant graph neural network (EGNN). There are two channels in MEG-PPIS: the original graph and the subgraph obtained by graph pooling. The model can iteratively update the features of the original graph and subgraph through the weight-sharing EGNN. Subsequently, the max-pooling operation aggregates the updated features of the original graph and subgraph. Ultimately, the model feeds node features into the prediction layer to obtain prediction results. Comparative assessments against other methods on benchmark datasets reveal that MEG-PPIS achieves optimal performance across all evaluation metrics and gets the fastest runtime. Furthermore, specific case studies demonstrate that our method can predict more true positive and true negative sites than the current best method, proving that our model achieves better performance in the PPIS prediction task. AVAILABILITY AND IMPLEMENTATION: The data and code are available at https://github.com/dhz234/MEG-PPIS.git.


Neural Networks, Computer , Protein Interaction Mapping , Protein Interaction Mapping/methods , Proteins/metabolism , Proteins/chemistry , Algorithms , Databases, Protein , Computational Biology/methods , Protein Interaction Maps
15.
Comput Methods Programs Biomed ; 250: 108188, 2024 Jun.
Article En | MEDLINE | ID: mdl-38657382

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.


Algorithms , Protein Interaction Mapping , Protein Interaction Mapping/methods , Protein Interaction Maps , Software , Sequence Alignment , Computational Biology/methods , Proteins/chemistry , Proteins/metabolism , Humans
16.
BMC Bioinformatics ; 25(1): 157, 2024 Apr 20.
Article En | MEDLINE | ID: mdl-38643108

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.


Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Protein Interaction Mapping/methods , Algorithms , Protein Interaction Maps , Computational Biology/methods
17.
Nat Commun ; 15(1): 3516, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664367

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.


Cross-Linking Reagents , Photometry , Protein Denaturation , Cross-Linking Reagents/chemistry , Photometry/methods , Proteins/chemistry , Proteins/metabolism , Electrophoresis, Polyacrylamide Gel/methods , Protein Interaction Mapping/methods , Mass Spectrometry/methods , Humans
18.
Methods Mol Biol ; 2787: 305-313, 2024.
Article En | MEDLINE | ID: mdl-38656499

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.


Arabidopsis , Plant Leaves , Protoplasts , Arabidopsis/metabolism , Arabidopsis/genetics , Protoplasts/metabolism , Plant Leaves/metabolism , Plant Leaves/genetics , Protein Interaction Mapping/methods , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Microscopy, Fluorescence/methods , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Nicotiana/metabolism , Nicotiana/genetics , Protein Binding , Agrobacterium/genetics , Agrobacterium/metabolism
19.
Methods Mol Biol ; 2806: 219-227, 2024.
Article En | MEDLINE | ID: mdl-38676806

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.


Immunoprecipitation , Protein Interaction Mapping , Immunoprecipitation/methods , Humans , Animals , Protein Interaction Mapping/methods , Mice , Protein Binding , Heterografts , Proteins/metabolism
20.
J Am Soc Mass Spectrom ; 35(5): 1055-1058, 2024 May 01.
Article En | MEDLINE | ID: mdl-38606722

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.


Proteomics , Software , Proteomics/methods , Mass Spectrometry/methods , Humans , Protein Interaction Mapping/methods , Biotinylation , Peptides/analysis , Peptides/chemistry , Peptides/metabolism
...