Статті в журналах з теми "Protein – protein interactions (PPI)"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Protein – protein interactions (PPI).

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-50 статей у журналах для дослідження на тему "Protein – protein interactions (PPI)".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте статті в журналах для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Kusova, Aleksandra M., Aleksandr E. Sitnitsky, Vladimir N. Uversky, and Yuriy F. Zuev. "Effect of Protein–Protein Interactions on Translational Diffusion of Spheroidal Proteins." International Journal of Molecular Sciences 23, no. 16 (August 17, 2022): 9240. http://dx.doi.org/10.3390/ijms23169240.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
One of the commonly accepted approaches to estimate protein–protein interactions (PPI) in aqueous solutions is the analysis of their translational diffusion. The present review article observes a phenomenological approach to analyze PPI effects via concentration dependencies of self- and collective translational diffusion coefficient for several spheroidal proteins derived from the pulsed field gradient NMR (PFG NMR) and dynamic light scattering (DLS), respectively. These proteins are rigid globular α-chymotrypsin (ChTr) and human serum albumin (HSA), and partly disordered α-casein (α-CN) and β-lactoglobulin (β-Lg). The PPI analysis enabled us to reveal the dominance of intermolecular repulsion at low ionic strength of solution (0.003–0.01 M) for all studied proteins. The increase in the ionic strength to 0.1–1.0 M leads to the screening of protein charges, resulting in the decrease of the protein electrostatic potential. The increase of the van der Waals potential for ChTr and α-CN characterizes their propensity towards unstable weak attractive interactions. The decrease of van der Waals interactions for β-Lg is probably associated with the formation of stable oligomers by this protein. The PPI, estimated with the help of interaction potential and idealized spherical molecular geometry, are in good agreement with experimental data.
2

CHUA, HON NIAN, KANG NING, WING-KIN SUNG, HON WAI LEONG, and LIMSOON WONG. "USING INDIRECT PROTEIN–PROTEIN INTERACTIONS FOR PROTEIN COMPLEX PREDICTION." Journal of Bioinformatics and Computational Biology 06, no. 03 (June 2008): 435–66. http://dx.doi.org/10.1142/s0219720008003497.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein complexes are fundamental for understanding principles of cellular organizations. As the sizes of protein–protein interaction (PPI) networks are increasing, accurate and fast protein complex prediction from these PPI networks can serve as a guide for biological experiments to discover novel protein complexes. However, it is not easy to predict protein complexes from PPI networks, especially in situations where the PPI network is noisy and still incomplete. Here, we study the use of indirect interactions between level-2 neighbors (level-2 interactions) for protein complex prediction. We know from previous work that proteins which do not interact but share interaction partners (level-2 neighbors) often share biological functions. We have proposed a method in which all direct and indirect interactions are first weighted using topological weight (FS-Weight), which estimates the strength of functional association. Interactions with low weight are removed from the network, while level-2 interactions with high weight are introduced into the interaction network. Existing clustering algorithms can then be applied to this modified network. We have also proposed a novel algorithm that searches for cliques in the modified network, and merge cliques to form clusters using a "partial clique merging" method. Experiments show that (1) the use of indirect interactions and topological weight to augment protein–protein interactions can be used to improve the precision of clusters predicted by various existing clustering algorithms; and (2) our complex-finding algorithm performs very well on interaction networks modified in this way. Since no other information except the original PPI network is used, our approach would be very useful for protein complex prediction, especially for prediction of novel protein complexes.
3

Abdullah, Syahid, Wisnu Ananta Kusuma, and Sony Hartono Wijaya. "Sequence-based prediction of protein-protein interaction using autocorrelation features and machine learning." Jurnal Teknologi dan Sistem Komputer 10, no. 1 (January 4, 2022): 1–11. http://dx.doi.org/10.14710/jtsiskom.2021.13984.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein-protein interaction (PPI) can define a protein's function by knowing the protein's position in a complex network of protein interactions. The number of PPIs that have been identified is relatively small. Therefore, several studies were conducted to predict PPI using protein sequence information. This research compares the performance of three autocorrelation methods: Moran, Geary, and Moreau-Broto, in extracting protein sequence features to predict PPI. The results of the three extractions are then applied to three machine learning algorithms, namely k-Nearest Neighbor (KNN), Random Forest, and Support Vector Machine (SVM). The prediction models with the three autocorrelation methods can produce predictions with high average accuracy, which is 95.34% for Geary in KNN, 97.43% for Geary in RF, and 97.11% for Geary and Moran in SVM. In addition, the interacting protein pairs tend to have similar autocorrelation characteristics. Thus, the autocorrelation method can be used to predict PPI well.
4

Dong, Yun Yuan, and Xian Chun Zhang. "Nonessential-Nonhub Proteins in the Protein-Protein Interaction Network." Advanced Materials Research 934 (May 2014): 159–64. http://dx.doi.org/10.4028/www.scientific.net/amr.934.159.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein-protein interaction (PPI) networks provide a simplified overview of the web of interactions that take place inside a cell. According to the centrality-lethality rule, hub proteins (proteins with high degree) tend to be essential in the PPI network. Moreover, there are also many low degree proteins in the PPI network, but they have different lethality. Some of them are essential proteins (essential-nonhub proteins), and the others are not (nonessential-nonhub proteins). In order to explain why nonessential-nonhub proteins don’t have essentiality, we propose a new measure n-iep (the number of essential neighbors) and compare nonessential-nonhub proteins with essential-nonhub proteins from topological, evolutionary and functional view. The comparison results show that there are statistical differences between nonessential-nonhub proteins and essential-nonhub proteins in centrality measures, clustering coefficient, evolutionary rate and the number of essential neighbors. These are reasons why nonessential-nonhub proteins don’t have lethality.
5

Poot Velez, Albros Hermes, Fernando Fontove, and Gabriel Del Rio. "Protein–Protein Interactions Efficiently Modeled by Residue Cluster Classes." International Journal of Molecular Sciences 21, no. 13 (July 6, 2020): 4787. http://dx.doi.org/10.3390/ijms21134787.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Predicting protein–protein interactions (PPI) represents an important challenge in structural bioinformatics. Current computational methods display different degrees of accuracy when predicting these interactions. Different factors were proposed to help improve these predictions, including choosing the proper descriptors of proteins to represent these interactions, among others. In the current work, we provide a representative protein structure that is amenable to PPI classification using machine learning approaches, referred to as residue cluster classes. Through sampling and optimization, we identified the best algorithm–parameter pair to classify PPI from more than 360 different training sets. We tested these classifiers against PPI datasets that were not included in the training set but shared sequence similarity with proteins in the training set to reproduce the situation of most proteins sharing sequence similarity with others. We identified a model with almost no PPI error (96–99% of correctly classified instances) and showed that residue cluster classes of protein pairs displayed a distinct pattern between positive and negative protein interactions. Our results indicated that residue cluster classes are structural features relevant to model PPI and provide a novel tool to mathematically model the protein structure/function relationship.
6

Orasch, Oliver, Noah Weber, Michael Müller, Amir Amanzadi, Chiara Gasbarri, and Christopher Trummer. "Protein–Protein Interaction Prediction for Targeted Protein Degradation." International Journal of Molecular Sciences 23, no. 13 (June 24, 2022): 7033. http://dx.doi.org/10.3390/ijms23137033.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein–protein interactions (PPIs) play a fundamental role in various biological functions; thus, detecting PPI sites is essential for understanding diseases and developing new drugs. PPI prediction is of particular relevance for the development of drugs employing targeted protein degradation, as their efficacy relies on the formation of a stable ternary complex involving two proteins. However, experimental methods to detect PPI sites are both costly and time-intensive. In recent years, machine learning-based methods have been developed as screening tools. While they are computationally more efficient than traditional docking methods and thus allow rapid execution, these tools have so far primarily been based on sequence information, and they are therefore limited in their ability to address spatial requirements. In addition, they have to date not been applied to targeted protein degradation. Here, we present a new deep learning architecture based on the concept of graph representation learning that can predict interaction sites and interactions of proteins based on their surface representations. We demonstrate that our model reaches state-of-the-art performance using AUROC scores on the established MaSIF dataset. We furthermore introduce a new dataset with more diverse protein interactions and show that our model generalizes well to this new data. These generalization capabilities allow our model to predict the PPIs relevant for targeted protein degradation, which we show by demonstrating the high accuracy of our model for PPI prediction on the available ternary complex data. Our results suggest that PPI prediction models can be a valuable tool for screening protein pairs while developing new drugs for targeted protein degradation.
7

Velasco-García, Roberto, and Rocío Vargas-Martínez. "The study of protein–protein interactions in bacteria." Canadian Journal of Microbiology 58, no. 11 (November 2012): 1241–57. http://dx.doi.org/10.1139/w2012-104.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Many of the functions fulfilled by proteins in the cell require specific protein–protein interactions (PPI). During the last decade, the use of high-throughput experimental technologies, primarily based on the yeast 2-hybrid system, generated extensive data currently located in public databases. This information has been used to build interaction networks for different species. Unfortunately, due to the nature of the yeast 2-hybrid system, these databases contain many false positives and negatives, thus they require purging. A method for confirming these PPI is to test them using a technique that operates in vivo and detects binary PPI. This article comprises an overview of the study of PPI and describes the main techniques that have been used to identify bacterial PPI, prioritizing those that can be used for their verification, and it also mentions a number of PPI that have been identified or confirmed using these methods.
8

Ban Bolly, Hendrikus Masang, Yulius Hermanto, Ahmad Faried, Muhammad Zafrullah Arifin, Trajanus Laurens Yembise, and Firman Fuad Wirakusumah. "Protein-protein Interaction Analysis of Contributing Molecules in Dura mater Healing Process." International Journal of ChemTech Research 13, no. 3 (2020): 73–82. http://dx.doi.org/10.20902/jctr.2019.130302.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Background: Dura mater is a special tissue that fulfills a critical function in brain anatomy and physiology. This tissue contains numerous cells, stem cells, and growth factors. This research investigates the protein interaction contributing to dura mater healing process. Methods: We use the available analysis software to perform the protein-protein interaction (PPI) analysis (http://gpsprot.org/index.php). GPS Protein is an interactive platform for visualizing human protein interaction by integrating HIPPIE and CORUM databases. We excluded HIV-1 proteomic and RNAi databases, instead focused on human PPI (Confidence level 0.75). Two proteins were inputted as query to identify the potential protein network in Dura mater healing according to previous studies, i.e. fibroblast growth factor-2 (FGF2) and transforming growth factor beta-1 (TGFβ1). Results: PPI results shows a high level (confidence level > 0.75) of protein-protein interaction of TGFβ1 to 197 other proteins (Confidence level ranges: 0.49 - 0.87), and PPI of FGF2 to 26 other proteins (Confidence level ranges: 0.0-0.97). TGFβ1 interactions showed the important interactions to some remodeling proteins. TGFβ1 encoded regulates cell proliferation, differentiation, growth, expression modulation and the activation of other growth factors. It also induces epithelial-to-mesenchymal transition (EMT) and cell migration. Conclusion: This bioinformatics approach is the more efficient and cheaper method for analyzing the molecular aspect of protein that has a special contribution in Dura mater healing process. These results could beneficial in focusing further researches for more complex laboratory examinations.
9

Kaur, Rajpreet, Poonam Khullar, and Anita Gupta. "Protein-Protein Interactions Followed by in-Situ Synthesis of Gold Nanoparticles." ECS Transactions 107, no. 1 (April 24, 2022): 16375–90. http://dx.doi.org/10.1149/10701.16375ecst.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein-protein interaction (PPI) plays an important role in various biochemical processes. Various binary mixtures of industrially important zein have been blended with rice and wheat proteins to expand their biological applicability. Zein-rice, zein-hard wheat, and zein-soft wheat mixtures show different behavior toward in vitro synthesis of Au NPs. This can also be explained by the TEM, FESEM, and DLS studies of PPI-Au NPs mixtures. Zein-rice-Au NPs mixture showed their mutual favorable interaction with high colloidal stability and high reduction ability towards nucleating Au NPs. This may be due to both hydrophobic and hydrophilic interaction of zein-rice PPI, which resulted in regular and shape-controlled Au NPs. In the case of zein-wheat PPI interaction, lesser hydrophobic interaction resulted in the incomplete unfolding of wheat protein and demixing of wheat protein with zein protein to anisotropic and irregular Au NPs. The study of these interactions will lead to the development of therapeutic centers toward different diseases and their environmental applications.
10

Yang, Lei, and Xianglong Tang. "Protein-Protein Interactions Prediction Based on Iterative Clique Extension with Gene Ontology Filtering." Scientific World Journal 2014 (2014): 1–6. http://dx.doi.org/10.1155/2014/523634.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cliques (maximal complete subnets) in protein-protein interaction (PPI) network are an important resource used to analyze protein complexes and functional modules. Clique-based methods of predicting PPI complement the data defection from biological experiments. However, clique-based predicting methods only depend on the topology of network. The false-positive and false-negative interactions in a network usually interfere with prediction. Therefore, we propose a method combining clique-based method of prediction and gene ontology (GO) annotations to overcome the shortcoming and improve the accuracy of predictions. According to different GO correcting rules, we generate two predicted interaction sets which guarantee the quality and quantity of predicted protein interactions. The proposed method is applied to the PPI network from the Database of Interacting Proteins (DIP) and most of the predicted interactions are verified by another biological database, BioGRID. The predicted protein interactions are appended to the original protein network, which leads to clique extension and shows the significance of biological meaning.
11

Zhang, Jinxiong, Cheng Zhong, Hai Xiang Lin, and Mian Wang. "Identifying Protein Complexes from Dynamic Temporal Interval Protein-Protein Interaction Networks." BioMed Research International 2019 (August 21, 2019): 1–17. http://dx.doi.org/10.1155/2019/3726721.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Identification of protein complex is very important for revealing the underlying mechanism of biological processes. Many computational methods have been developed to identify protein complexes from static protein-protein interaction (PPI) networks. Recently, researchers are considering the dynamics of protein-protein interactions. Dynamic PPI networks are closer to reality in the cell system. It is expected that more protein complexes can be accurately identified from dynamic PPI networks. In this paper, we use the undulating degree above the base level of gene expression instead of the gene expression level to construct dynamic temporal PPI networks. Further we convert dynamic temporal PPI networks into dynamic Temporal Interval Protein Interaction Networks (TI-PINs) and propose a novel method to accurately identify more protein complexes from the constructed TI-PINs. Owing to preserving continuous interactions within temporal interval, the constructed TI-PINs contain more dynamical information for accurately identifying more protein complexes. Our proposed identification method uses multisource biological data to judge whether the joint colocalization condition, the joint coexpression condition, and the expanding cluster condition are satisfied; this is to ensure that the identified protein complexes have the features of colocalization, coexpression, and functional homogeneity. The experimental results on yeast data sets demonstrated that using the constructed TI-PINs can obtain better identification of protein complexes than five existing dynamic PPI networks, and our proposed identification method can find more protein complexes accurately than four other methods.
12

Klein, Mark. "Targeting Protein-Protein Interactions to Inhibit Cyclin-Dependent Kinases." Pharmaceuticals 16, no. 4 (March 31, 2023): 519. http://dx.doi.org/10.3390/ph16040519.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cyclin-dependent kinases (CDKs) play diverse and critical roles in normal cells and may be exploited as targets in cancer therapeutic strategies. CDK4 inhibitors are currently approved for treatment in advanced breast cancer. This success has led to continued pursuit of targeting other CDKs. One challenge has been in the development of inhibitors that are highly selective for individual CDKs as the ATP-binding site is highly conserved across this family of proteins. Protein-protein interactions (PPI) tend to have less conservation amongst different proteins, even within protein families, making targeting PPI an attractive approach to improving drug selectivity. However, PPI can be challenging to target due to structural and physicochemical features of these interactions. A review of the literature specific to studies focused on targeting PPI involving CDKs 2, 4, 5, and 9 was conducted and is presented here. Promising lead molecules to target select CDKs have been discovered. None of the lead molecules discovered have led to FDA approval; however, the studies covered in this review lay the foundation for further discovery and develop of PPI inhibitors for CDKs.
13

Dallago, Christian, Tatyana Goldberg, Miguel Angel Andrade-Navarro, Gregorio Alanis-Lobato, and Burkhard Rost. "CellMap visualizes protein-protein interactions and subcellular localization." F1000Research 6 (October 11, 2017): 1824. http://dx.doi.org/10.12688/f1000research.12707.1.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Many tools visualize protein-protein interaction (PPI) networks. The tool introduced here, CellMap, adds one crucial novelty by visualizing PPI networks in the context of subcellular localization, i.e. the location in the cell or cellular component in which a PPI happens. Users can upload images of cells and define areas of interest against which PPIs for selected proteins are displayed (by default on a cartoon of a cell). Annotations of localization are provided by the user or through our in-house database. The visualizer and server are written in JavaScript, making CellMap easy to customize and to extend by researchers and developers.
14

Dallago, Christian, Tatyana Goldberg, Miguel Angel Andrade-Navarro, Gregorio Alanis-Lobato, and Burkhard Rost. "CellMap visualizes protein-protein interactions and subcellular localization." F1000Research 6 (February 1, 2018): 1824. http://dx.doi.org/10.12688/f1000research.12707.2.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Many tools visualize protein-protein interaction (PPI) networks. The tool introduced here, CellMap, adds one crucial novelty by visualizing PPI networks in the context of subcellular localization, i.e. the location in the cell or cellular component in which a PPI happens. Users can upload images of cells and define areas of interest against which PPIs for selected proteins are displayed (by default on a cartoon of a cell). Annotations of localization are provided by the user or through our in-house database. The visualizer and server are written in JavaScript, making CellMap easy to customize and to extend by researchers and developers.
15

Blaszczak, Ewa, Natalia Lazarewicz, Aswani Sudevan, Robert Wysocki, and Gwenaël Rabut. "Protein-fragment complementation assays for large-scale analysis of protein–protein interactions." Biochemical Society Transactions 49, no. 3 (June 22, 2021): 1337–48. http://dx.doi.org/10.1042/bst20201058.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein–protein interactions (PPIs) orchestrate nearly all biological processes. They are also considered attractive drug targets for treating many human diseases, including cancers and neurodegenerative disorders. Protein-fragment complementation assays (PCAs) provide a direct and straightforward way to study PPIs in living cells or multicellular organisms. Importantly, PCAs can be used to detect the interaction of proteins expressed at endogenous levels in their native cellular environment. In this review, we present the principle of PCAs and discuss some of their advantages and limitations. We describe their application in large-scale experiments to investigate PPI networks and to screen or profile PPI targeting compounds.
16

Kazemi-Pour, Ali, Bahram Goliaei, and Hamid Pezeshk. "Protein Complex Discovery by Interaction Filtering from Protein Interaction Networks Using Mutual Rank Coexpression and Sequence Similarity." BioMed Research International 2015 (2015): 1–7. http://dx.doi.org/10.1155/2015/165186.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The evaluation of the biological networks is considered the essential key to understanding the complex biological systems. Meanwhile, the graph clustering algorithms are mostly used in the protein-protein interaction (PPI) network analysis. The complexes introduced by the clustering algorithms include noise proteins. The error rate of the noise proteins in the PPI network researches is about 40–90%. However, only 30–40% of the existing interactions in the PPI databases depend on the specific biological function. It is essential to eliminate the noise proteins and the interactions from the complexes created via clustering methods. We have introduced new methods of weighting interactions in protein clusters and the splicing of noise interactions and proteins-based interactions on their weights. The coexpression and the sequence similarity of each pair of proteins are considered the edge weight of the proteins in the network. The results showed that the edge filtering based on the amount of coexpression acts similar to the node filtering via graph-based characteristics. Regarding the removal of the noise edges, the edge filtering has a significant advantage over the graph-based method. The edge filtering based on the amount of sequence similarity has the ability to remove the noise proteins and the noise interactions.
17

Usman, Muhammad Syafiuddin, Wisnu Ananta Kusuma, Farit Mochamad Afendi, and Rudi Heryanto. "Identification of Significant Proteins Associated with Diabetes Mellitus Using Network Analysis of Protein-Protein Interactions." Computer Engineering and Applications Journal 8, no. 1 (February 1, 2019): 41–52. http://dx.doi.org/10.18495/comengapp.v8i1.283.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Computation approach to identify significance of proteins related with disease was proposed as one of the solutions from the problem of experimental method application which is generally high cost and time consuming. The case of study was conducted on Diabetes Melitus (DM) type 2 diseases. Identification of significant proteins was conducted by constructing protein-protein interactions network and then analyzing the network topology. Dataset was obtained from Online Mendelian Inheritance in Man (OMIM) and Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) which provided protein data related with a disease and Protein-Protein Interaction (PPI), respectively. The results of topology analysis towards Protein-Protein Interaction (PPI) showed that there were 21 significant protein associated with DM where INS as a network center protein and AKTI, TCF7L2, KCNJ11, PPARG, GCG, INSR, IAPP, SOCS3 were proteins that had close relation directly with INS.
18

Diansyah, Mohammad Romano, Wisnu Ananta Kusuma, and Annisa Annisa. "Identification of significant protein in protein-protein interaction of Alzheimer disease using top-k representative skyline query." Jurnal Teknologi dan Sistem Komputer 9, no. 3 (April 24, 2021): 126–32. http://dx.doi.org/10.14710/jtsiskom.2021.13985.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Alzheimer's disease is the most common neurodegenerative disease. This study aims to analyze protein-protein interaction (PPI) to provide a better understanding of multifactorial neurodegenerative diseases and can be used to find proteins that have a significant role in Alzheimer's disease. PPI data were obtained from experimental and computational predictions and analyzed using centrality measures. The Top-k RSP method was applied to find significant proteins in PPI networks using the dominance rule. The method was applied to the PPI data with the interaction sources from the experimental and experiment+prediction. The results indicate that APP and PSEN1 are significant proteins for Alzheimer's disease. This study also showed that both data sources (experiment+prediction) and the Top-k RSP algorithm proved useful for PPI analysis of Alzheimer's disease.
19

Alborzi, Seyed Ziaeddin, Amina Ahmed Nacer, Hiba Najjar, David W. Ritchie, and Marie-Dominique Devignes. "PPIDomainMiner: Inferring domain-domain interactions from multiple sources of protein-protein interactions." PLOS Computational Biology 17, no. 8 (August 9, 2021): e1008844. http://dx.doi.org/10.1371/journal.pcbi.1008844.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Many biological processes are mediated by protein-protein interactions (PPIs). Because protein domains are the building blocks of proteins, PPIs likely rely on domain-domain interactions (DDIs). Several attempts exist to infer DDIs from PPI networks but the produced datasets are heterogeneous and sometimes not accessible, while the PPI interactome data keeps growing. We describe a new computational approach called “PPIDM” (Protein-Protein Interactions Domain Miner) for inferring DDIs using multiple sources of PPIs. The approach is an extension of our previously described “CODAC” (Computational Discovery of Direct Associations using Common neighbors) method for inferring new edges in a tripartite graph. The PPIDM method has been applied to seven widely used PPI resources, using as “Gold-Standard” a set of DDIs extracted from 3D structural databases. Overall, PPIDM has produced a dataset of 84,552 non-redundant DDIs. Statistical significance (p-value) is calculated for each source of PPI and used to classify the PPIDM DDIs in Gold (9,175 DDIs), Silver (24,934 DDIs) and Bronze (50,443 DDIs) categories. Dataset comparison reveals that PPIDM has inferred from the 2017 releases of PPI sources about 46% of the DDIs present in the 2020 release of the 3did database, not counting the DDIs present in the Gold-Standard. The PPIDM dataset contains 10,229 DDIs that are consistent with more than 13,300 PPIs extracted from the IMEx database, and nearly 23,300 DDIs (27.5%) that are consistent with more than 214,000 human PPIs extracted from the STRING database. Examples of newly inferred DDIs covering more than 10 PPIs in the IMEx database are provided. Further exploitation of the PPIDM DDI reservoir includes the inventory of possible partners of a protein of interest and characterization of protein interactions at the domain level in combination with other methods. The result is publicly available at http://ppidm.loria.fr/.
20

Liu, Hongfang, Manabu Torii, Guixian Xu, and Johannes Goll. "Classification Systems for Bacterial Protein-Protein Interaction Document Retrieval." International Journal of Computational Models and Algorithms in Medicine 1, no. 1 (January 2010): 34–44. http://dx.doi.org/10.4018/jcmam.2010072003.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein-protein interaction (PPI) networks are essential to understand the fundamental processes governing cell biology. Recently, studying PPI networks becomes possible due to advances in experimental high-throughput genomics and proteomics technologies. Many interactions from such high-throughput studies and most interactions from small-scale studies are reported only in the scientific literature and thus are not accessible in a readily analyzable format. This has led to the birth of manual curation initiatives such as the International Molecular Exchange Consortium (IMEx). The manual curation of PPI knowledge can be accelerated by text mining systems to retrieve PPI-relevant articles (article retrieval) and extract PPI-relevant knowledge (information extraction). In this article, the authors focus on article retrieval and define the task as binary classification where PPI-relevant articles are positives and the others are negatives. In order to build such classifier, an annotated corpus is needed. It is very expensive to obtain an annotated corpus manually but a noisy and imbalanced annotated corpus can be obtained automatically, where a collection of positive documents can be retrieved from existing PPI knowledge bases and a large number of unlabeled documents (most of them are negatives) can be retrieved from PubMed. They compared the performance of several machine learning algorithms by varying the ratio of the number of positives to the number of unlabeled documents and the number of features used.
21

Idrees, Sobia, Åsa Pérez-Bercoff, and Richard J. Edwards. "SLiM-Enrich: computational assessment of protein–protein interaction data as a source of domain-motif interactions." PeerJ 6 (October 31, 2018): e5858. http://dx.doi.org/10.7717/peerj.5858.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Many important cellular processes involve protein–protein interactions (PPIs) mediated by a Short Linear Motif (SLiM) in one protein interacting with a globular domain in another. Despite their significance, these domain-motif interactions (DMIs) are typically low affinity, which makes them challenging to identify by classical experimental approaches, such as affinity pulldown mass spectrometry (AP-MS) and yeast two-hybrid (Y2H). DMIs are generally underrepresented in PPI networks as a result. A number of computational methods now exist to predict SLiMs and/or DMIs from experimental interaction data but it is yet to be established how effective different PPI detection methods are for capturing these low affinity SLiM-mediated interactions. Here, we introduce a new computational pipeline (SLiM-Enrich) to assess how well a given source of PPI data captures DMIs and thus, by inference, how useful that data should be for SLiM discovery. SLiM-Enrich interrogates a PPI network for pairs of interacting proteins in which the first protein is known or predicted to interact with the second protein via a DMI. Permutation tests compare the number of known/predicted DMIs to the expected distribution if the two sets of proteins are randomly associated. This provides an estimate of DMI enrichment within the data and the false positive rate for individual DMIs. As a case study, we detect significant DMI enrichment in a high-throughput Y2H human PPI study. SLiM-Enrich analysis supports Y2H data as a source of DMIs and highlights the high false positive rates associated with naïve DMI prediction. SLiM-Enrich is available as an R Shiny app. The code is open source and available via a GNU GPL v3 license at: https://github.com/slimsuite/SLiMEnrich. A web server is available at: http://shiny.slimsuite.unsw.edu.au/SLiMEnrich/.
22

Jung, Dongmin, and Xijin Ge. "PPInfer: a Bioconductor package for inferring functionally related proteins using protein interaction networks." F1000Research 6 (November 7, 2017): 1969. http://dx.doi.org/10.12688/f1000research.12947.1.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Interactions between proteins occur in many, if not most, biological processes. This fact has motivated the development of a variety of experimental methods for the identification of protein-protein interaction (PPI) networks. Leveraging PPI data available STRING database, we use network-based statistical learning methods to infer the putative functions of proteins from the known functions of neighboring proteins on a PPI network. This package identifies such proteins often involved in the same or similar biological functions. The package is freely available at the Bioconductor web site (http://bioconductor.org/packages/PPInfer/).
23

Jung, Dongmin, and Xijin Ge. "PPInfer: a Bioconductor package for inferring functionally related proteins using protein interaction networks." F1000Research 6 (December 8, 2017): 1969. http://dx.doi.org/10.12688/f1000research.12947.2.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Interactions between proteins occur in many, if not most, biological processes. This fact has motivated the development of a variety of experimental methods for the identification of protein-protein interaction (PPI) networks. Leveraging PPI data available STRING database, we use network-based statistical learning methods to infer the putative functions of proteins from the known functions of neighboring proteins on a PPI network. This package identifies such proteins often involved in the same or similar biological functions. The package is freely available at the Bioconductor web site (http://bioconductor.org/packages/PPInfer/).
24

Jung, Dongmin, and Xijin Ge. "PPInfer: a Bioconductor package for inferring functionally related proteins using protein interaction networks." F1000Research 6 (March 12, 2018): 1969. http://dx.doi.org/10.12688/f1000research.12947.3.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Interactions between proteins occur in many, if not most, biological processes. This fact has motivated the development of a variety of experimental methods for the identification of protein-protein interaction (PPI) networks. Leveraging PPI data available in the STRING database, we use a network-based statistical learning methods to infer the putative functions of proteins from the known functions of neighboring proteins on a PPI network. This package identifies such proteins often involved in the same or similar biological functions. The package is freely available at the Bioconductor web site (http://bioconductor.org/packages/PPInfer/).
25

Dash, Radha Charan, and Kyle Hadden. "Protein–Protein Interactions in Translesion Synthesis." Molecules 26, no. 18 (September 13, 2021): 5544. http://dx.doi.org/10.3390/molecules26185544.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Translesion synthesis (TLS) is an error-prone DNA damage tolerance mechanism used by actively replicating cells to copy past DNA lesions and extend the primer strand. TLS ensures that cells continue replication in the presence of damaged DNA bases, albeit at the expense of an increased mutation rate. Recent studies have demonstrated a clear role for TLS in rescuing cancer cells treated with first-line genotoxic agents by allowing them to replicate and survive in the presence of chemotherapy-induced DNA lesions. The importance of TLS in both the initial response to chemotherapy and the long-term development of acquired resistance has allowed it to emerge as an interesting target for small molecule drug discovery. Proper TLS function is a complicated process involving a heteroprotein complex that mediates multiple attachment and switching steps through several protein–protein interactions (PPIs). In this review, we briefly describe the importance of TLS in cancer and provide an in-depth analysis of key TLS PPIs, focusing on key structural features at the PPI interface while also exploring the potential druggability of each key PPI.
26

Milano, Marianna, Giuseppe Agapito, and Mario Cannataro. "Challenges and Limitations of Biological Network Analysis." BioTech 11, no. 3 (July 7, 2022): 24. http://dx.doi.org/10.3390/biotech11030024.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
High-Throughput technologies are producing an increasing volume of data that needs large amounts of data storage, effective data models and efficient, possibly parallel analysis algorithms. Pathway and interactomics data are represented as graphs and add a new dimension of analysis, allowing, among other features, graph-based comparison of organisms’ properties. For instance, in biological pathway representation, the nodes can represent proteins, RNA and fat molecules, while the edges represent the interaction between molecules. Otherwise, biological networks such as Protein–Protein Interaction (PPI) Networks, represent the biochemical interactions among proteins by using nodes that model the proteins from a given organism, and edges that model the protein–protein interactions, whereas pathway networks enable the representation of biochemical-reaction cascades that happen within the cells or tissues. In this paper, we discuss the main models for standard representation of pathways and PPI networks, the data models for the representation and exchange of pathway and protein interaction data, the main databases in which they are stored and the alignment algorithms for the comparison of pathways and PPI networks of different organisms. Finally, we discuss the challenges and the limitations of pathways and PPI network representation and analysis. We have identified that network alignment presents a lot of open problems worthy of further investigation, especially concerning pathway alignment.
27

Winkler, Joanna, Evelien Mylle, Andreas De Meyer, Benjamin Pavie, Julie Merchie, Peter Grones, and Dani�l Van Damme. "Visualizing protein–protein interactions in plants by rapamycin-dependent delocalization." Plant Cell 33, no. 4 (January 25, 2021): 1101–17. http://dx.doi.org/10.1093/plcell/koab004.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Identifying protein–protein interactions (PPIs) is crucial for understanding biological processes. Many PPI tools are available, yet only some function within the context of a plant cell. Narrowing down even further, only a few tools allow complex multi-protein interactions to be visualized. Here, we present a conditional in vivo PPI tool for plant research that meets these criteria. Knocksideways in plants (KSP) is based on the ability of rapamycin to alter the localization of a bait protein and its interactors via the heterodimerization of FKBP and FRB domains. KSP is inherently free from many limitations of other PPI systems. This in vivo tool does not require spatial proximity of the bait and prey fluorophores and it is compatible with a broad range of fluorophores. KSP is also a conditional tool and therefore the visualization of the proteins in the absence of rapamycin acts as an internal control. We used KSP to confirm previously identified interactions in Nicotiana benthamiana leaf epidermal cells. Furthermore, the scripts that we generated allow the interactions to be quantified at high throughput. Finally, we demonstrate that KSP can easily be used to visualize complex multi-protein interactions. KSP is therefore a versatile tool with unique characteristics and applications that complements other plant PPI methods.
28

Gopalakrishnan, Sathyanarayanan, and Swaminathan Venkatraman. "Prediction of influential proteins and enzymes of certain diseases using a directed unimodular hypergraph." Mathematical Biosciences and Engineering 21, no. 1 (2023): 325–45. http://dx.doi.org/10.3934/mbe.2024015.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
<abstract><p>Protein-protein interaction (PPI) analysis based on mathematical modeling is an efficient means of identifying hub proteins, corresponding enzymes and many underlying structures. In this paper, a method for the analysis of PPI is introduced and used to analyze protein interactions of diseases such as Parkinson's, COVID-19 and diabetes melitus. A directed hypergraph is used to represent PPI interactions. A novel directed hypergraph depth-first search algorithm is introduced to find the longest paths. The minor hypergraph reduces the dimension of the directed hypergraph, representing the longest paths and results in the unimodular hypergraph. The property of unimodular hypergraph clusters influential proteins and enzymes that are related thereby providing potential avenues for disease treatment.</p></abstract>
29

Hu, Yang, Ying Zhang, Jun Ren, Yadong Wang, Zhenzhen Wang, and Jun Zhang. "Statistical Approaches for the Construction and Interpretation of Human Protein-Protein Interaction Network." BioMed Research International 2016 (2016): 1–7. http://dx.doi.org/10.1155/2016/5313050.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The overall goal is to establish a reliable human protein-protein interaction network and develop computational tools to characterize a protein-protein interaction (PPI) network and the role of individual proteins in the context of the network topology and their expression status. A novel and unique feature of our approach is that we assigned confidence measure to each derived interacting pair and account for the confidence in our network analysis. We integrated experimental data to infer human PPI network. Our model treated the true interacting status (yes versus no) for any given pair of human proteins as a latent variable whose value was not observed. The experimental data were the manifestation of interacting status, which provided evidence as to the likelihood of the interaction. The confidence of interactions would depend on the strength and consistency of the evidence.
30

IVANOV, ALEXIS S., OKSANA V. GNEDENKO, ANDREY A. MOLNAR, YURY V. MEZENTSEV, ANDREY V. LISITSA, and ALEXANDER I. ARCHAKOV. "PROTEIN–PROTEIN INTERACTIONS AS NEW TARGETS FOR DRUG DESIGN: VIRTUAL AND EXPERIMENTAL APPROACHES." Journal of Bioinformatics and Computational Biology 05, no. 02b (April 2007): 579–92. http://dx.doi.org/10.1142/s0219720007002825.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein–protein and protein–ligand interactions play a central role in biochemical reactions, and understanding these processes is an important task in different fields of biomedical science and drug discovery. Proteins often work in complex assemblies of several macromolecules and small ligands. The structural and functional description of protein–protein interactions (PPI) is very important for basic-, as well as applied research. The interface areas of protein complexes have unique structure and properties, so PPI represent prospective targets for a new generation of drugs. One of the key targets of PPI inhibitors are oligomeric enzymes. This report shows interactive links between virtual and experimental approaches in a total pipeline "from gene to drug" and using Surface Plasmon Resonance technology for experimentally assessing PPI. Our research is conducted on two oligomeric enzymes — HIV-1 protease (HIVp) (homo-dimer) and bacterial L-asparaginase (homo-tetramer). Using methods of molecular modeling and computational alanine scanning we obtained structural and functional description of PPI in these two enzymes. We also presented a real example of application of integral approach in searching inhibitors of HIVp dimerization — from virtual database mining up to experimental testing of lead compounds.
31

You, Zhu-Hong, Shuai Li, Xin Gao, Xin Luo, and Zhen Ji. "Large-Scale Protein-Protein Interactions Detection by Integrating Big Biosensing Data with Computational Model." BioMed Research International 2014 (2014): 1–9. http://dx.doi.org/10.1155/2014/598129.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein-protein interactions are the basis of biological functions, and studying these interactions on a molecular level is of crucial importance for understanding the functionality of a living cell. During the past decade, biosensors have emerged as an important tool for the high-throughput identification of proteins and their interactions. However, the high-throughput experimental methods for identifying PPIs are both time-consuming and expensive. On the other hand, high-throughput PPI data are often associated with high false-positive and high false-negative rates. Targeting at these problems, we propose a method for PPI detection by integrating biosensor-based PPI data with a novel computational model. This method was developed based on the algorithm of extreme learning machine combined with a novel representation of protein sequence descriptor. When performed on the large-scale human protein interaction dataset, the proposed method achieved 84.8% prediction accuracy with 84.08% sensitivity at the specificity of 85.53%. We conducted more extensive experiments to compare the proposed method with the state-of-the-art techniques, support vector machine. The achieved results demonstrate that our approach is very promising for detecting new PPIs, and it can be a helpful supplement for biosensor-based PPI data detection.
32

Chasapis, Christos T., Konstantinos Kelaidonis, Harry Ridgway, Vasso Apostolopoulos, and John M. Matsoukas. "The Human Myelin Proteome and Sub-Metalloproteome Interaction Map: Relevance to Myelin-Related Neurological Diseases." Brain Sciences 12, no. 4 (March 24, 2022): 434. http://dx.doi.org/10.3390/brainsci12040434.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Myelin in humans is composed of about 80% lipids and 20% protein. Initially, myelin protein composition was considered low, but various recent proteome analyses have identified additional myelin proteins. Although, the myelin proteome is qualitatively and quantitatively identified through complementary proteomic approaches, the corresponding Protein–Protein Interaction (PPI) network of myelin is not yet available. In the present work, the PPI network was constructed based on available experimentally supported protein interactions of myelin in PPI databases. The network comprised 2017 PPIs between 567 myelin proteins. Interestingly, structure-based in silico analysis revealed that 20% of the myelin proteins that are interconnected in the proposed PPI network are metal-binding proteins/enzymes that construct the main sub-PPI network of myelin proteome. Finally, the PPI networks of the myelin proteome and sub-metalloproteome were analyzed ontologically to identify the biochemical processes of the myelin proteins and the interconnectivity of myelin-associated diseases in the interactomes. The presented PPI dataset could provide a useful resource to the scientific community to further our understanding of human myelin biology and serve as a basis for future studies of myelin-related neurological diseases and particular autoimmune diseases such as multiple sclerosis where myelin epitopes are implicated.
33

Xu, Da, Hanxiao Xu, Yusen Zhang, Wei Chen, and Rui Gao. "Protein-Protein Interactions Prediction Based on Graph Energy and Protein Sequence Information." Molecules 25, no. 8 (April 16, 2020): 1841. http://dx.doi.org/10.3390/molecules25081841.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Identification of protein-protein interactions (PPIs) plays an essential role in the understanding of protein functions and cellular biological activities. However, the traditional experiment-based methods are time-consuming and laborious. Therefore, developing new reliable computational approaches has great practical significance for the identification of PPIs. In this paper, a novel prediction method is proposed for predicting PPIs using graph energy, named PPI-GE. Particularly, in the process of feature extraction, we designed two new feature extraction methods, the physicochemical graph energy based on the ionization equilibrium constant and isoelectric point and the contact graph energy based on the contact information of amino acids. The dipeptide composition method was used for order information of amino acids. After multi-information fusion, principal component analysis (PCA) was implemented for eliminating noise and a robust weighted sparse representation-based classification (WSRC) classifier was applied for sample classification. The prediction accuracies based on the five-fold cross-validation of the human, Helicobacter pylori (H. pylori), and yeast data sets were 99.49%, 97.15%, and 99.56%, respectively. In addition, in five independent data sets and two significant PPI networks, the comparative experimental results also demonstrate that PPI-GE obtained better performance than the compared methods.
34

Larsen, Peter E., Frank Collart, and Yang Dai. "Incorporating Network Topology Improves Prediction of Protein Interaction Networks from Transcriptomic Data." International Journal of Knowledge Discovery in Bioinformatics 1, no. 3 (July 2010): 1–19. http://dx.doi.org/10.4018/jkdb.2010070101.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The reconstruction of protein-protein interaction (PPI) networks from high-throughput experimental data is one of the most challenging problems in bioinformatics. These biological networks have specific topologies defined by the functional and evolutionary relationships between the proteins and the physical limitations imposed on proteins interacting in the three-dimensional space. In this paper, the authors propose a novel approach for the identification of potential protein-protein interactions based on the integration of known PPI network topology and transcriptomic data. The proposed method, Function Restricted Value Neighborhood (FRV-N), was used to reconstruct PPI networks using an experimental data set consisting of 170 yeast microarray profiles. The results of this analysis demonstrate that incorporating knowledge of interactome topology improves the ability of transcriptome analysis to reconstruct interaction networks with a high degree of biological relevance.
35

Nezamuldeen, Leena, and Mohsin Saleet Jafri. "Protein–Protein Interaction Network Extraction Using Text Mining Methods Adds Insight into Autism Spectrum Disorder." Biology 12, no. 10 (October 18, 2023): 1344. http://dx.doi.org/10.3390/biology12101344.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Text mining methods are being developed to assimilate the volume of biomedical textual materials that are continually expanding. Understanding protein–protein interaction (PPI) deficits would assist in explaining the genesis of diseases. In this study, we designed an automated system to extract PPIs from the biomedical literature that uses a deep learning sentence classification model, a pretrained word embedding, and a BiLSTM recurrent neural network with additional layers, a conditional random field (CRF) named entity recognition (NER) model, and shortest-dependency path (SDP) model using the SpaCy library in Python. The automated system ensures that it targets sentences that contain PPIs and not just these proteins mentioned in the framework of disease discovery or other context. Our first model achieved 13% greater precision on the Aimed/BioInfr benchmark corpus than the previous state-of-the-art BiLSTM neural network models. The NER model presented in this study achieved 98% precision on the Aimed/BioInfr corpus over previous models. In order to facilitate the production of an accurate representation of the PPI network, the processes were developed to systematically map the protein interactions in the texts. Overall, evaluating our system through the use of 6027 abstracts pertaining to seven proteins associated with Autism Spectrum Disorder completed the manually curated PPI network for these proteins. When it comes to complicated diseases, these networks would assist in understanding how PPI deficits contribute to disease development while also emphasizing the influence of interactions on protein function and biological processes.
36

Gemovic, Branislava, Neven Sumonja, Radoslav Davidovic, Vladimir Perovic, and Nevena Veljkovic. "Mapping of Protein-Protein Interactions: Web-Based Resources for Revealing Interactomes." Current Medicinal Chemistry 26, no. 21 (September 19, 2019): 3890–910. http://dx.doi.org/10.2174/0929867325666180214113704.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Background: The significant number of protein-protein interactions (PPIs) discovered by harnessing concomitant advances in the fields of sequencing, crystallography, spectrometry and two-hybrid screening suggests astonishing prospects for remodelling drug discovery. The PPI space which includes up to 650 000 entities is a remarkable reservoir of potential therapeutic targets for every human disease. In order to allow modern drug discovery programs to leverage this, we should be able to discern complete PPI maps associated with a specific disorder and corresponding normal physiology. Objective: Here, we will review community available computational programs for predicting PPIs and web-based resources for storing experimentally annotated interactions. Methods: We compared the capacities of prediction tools: iLoops, Struck2Net, HOMCOS, COTH, PrePPI, InterPreTS and PRISM to predict recently discovered protein interactions. Results: We described sequence-based and structure-based PPI prediction tools and addressed their peculiarities. Additionally, since the usefulness of prediction algorithms critically depends on the quality and quantity of the experimental data they are built on; we extensively discussed community resources for protein interactions. We focused on the active and recently updated primary and secondary PPI databases, repositories specialized to the subject or species, as well as databases that include both experimental and predicted PPIs. Conclusion: PPI complexes are the basis of important physiological processes and therefore, possible targets for cell-penetrating ligands. Reliable computational PPI predictions can speed up new target discoveries through prioritization of therapeutically relevant protein–protein complexes for experimental studies.
37

Wang, Derui, and Jingyu Hou. "Explore the hidden treasure in protein–protein interaction networks — An iterative model for predicting protein functions." Journal of Bioinformatics and Computational Biology 13, no. 05 (October 2015): 1550026. http://dx.doi.org/10.1142/s0219720015500262.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein–protein interaction networks constructed by high throughput technologies provide opportunities for predicting protein functions. A lot of approaches and algorithms have been applied on PPI networks to predict functions of unannotated proteins over recent decades. However, most of existing algorithms and approaches do not consider unannotated proteins and their corresponding interactions in the prediction process. On the other hand, algorithms which make use of unannotated proteins have limited prediction performance. Moreover, current algorithms are usually one-off predictions. In this paper, we propose an iterative approach that utilizes unannotated proteins and their interactions in prediction. We conducted experiments to evaluate the performance and robustness of the proposed iterative approach. The iterative approach maximally improved the prediction performance by 50%–80% when there was a high proportion of unannotated neighborhood protein in the network. The iterative approach also showed robustness in various types of protein interaction network. Importantly, our iterative approach initially proposes an idea that iteratively incorporates the interaction information of unannotated proteins into the protein function prediction and can be applied on existing prediction algorithms to improve prediction performance.
38

El Khamlichi, Chayma, Flora Reverchon-Assadi, Nadège Hervouet-Coste, Lauren Blot, Eric Reiter, and Séverine Morisset-Lopez. "Bioluminescence Resonance Energy Transfer as a Method to Study Protein-Protein Interactions: Application to G Protein Coupled Receptor Biology." Molecules 24, no. 3 (February 1, 2019): 537. http://dx.doi.org/10.3390/molecules24030537.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The bioluminescence resonance energy transfer (BRET) approach involves resonance energy transfer between a light-emitting enzyme and fluorescent acceptors. The major advantage of this technique over biochemical methods is that protein-protein interactions (PPI) can be monitored without disrupting the natural environment, frequently altered by detergents and membrane preparations. Thus, it is considered as one of the most versatile technique for studying molecular interactions in living cells at “physiological” expression levels. BRET analysis has been applied to study many transmembrane receptor classes including G-protein coupled receptors (GPCR). It is well established that these receptors may function as dimeric/oligomeric forms and interact with multiple effectors to transduce the signal. Therefore, they are considered as attractive targets to identify PPI modulators. In this review, we present an overview of the different BRET systems developed up to now and their relevance to identify inhibitors/modulators of protein–protein interaction. Then, we introduce the different classes of agents that have been recently developed to target PPI, and provide some examples illustrating the use of BRET-based assays to identify and characterize innovative PPI modulators in the field of GPCRs biology. Finally, we discuss the main advantages and the limits of BRET approach to characterize PPI modulators.
39

Ohue, Masahito, Yuki Kojima, and Takatsugu Kosugi. "Generating Potential Protein-Protein Interaction Inhibitor Molecules Based on Physicochemical Properties." Molecules 28, no. 15 (July 26, 2023): 5652. http://dx.doi.org/10.3390/molecules28155652.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein-protein interactions (PPIs) are associated with various diseases; hence, they are important targets in drug discovery. However, the physicochemical empirical properties of PPI-targeted drugs are distinct from those of conventional small molecule oral pharmaceuticals, which adhere to the ”rule of five (RO5)”. Therefore, developing PPI-targeted drugs using conventional methods, such as molecular generation models, is challenging. In this study, we propose a molecular generation model based on deep reinforcement learning that is specialized for the production of PPI inhibitors. By introducing a scoring function that can represent the properties of PPI inhibitors, we successfully generated potential PPI inhibitor compounds. These newly constructed virtual compounds possess the desired properties for PPI inhibitors, and they show similarity to commercially available PPI libraries. The virtual compounds are freely available as a virtual library.
40

Ghedira, Kais, Yosr Hamdi, Abir El Béji, and Houcemeddine Othman. "An Integrative Computational Approach for the Prediction of Human-Plasmodium Protein-Protein Interactions." BioMed Research International 2020 (December 19, 2020): 1–11. http://dx.doi.org/10.1155/2020/2082540.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Host-pathogen molecular cross-talks are critical in determining the pathophysiology of a specific infection. Most of these cross-talks are mediated via protein-protein interactions between the host and the pathogen (HP-PPI). Thus, it is essential to know how some pathogens interact with their hosts to understand the mechanism of infections. Malaria is a life-threatening disease caused by an obligate intracellular parasite belonging to the Plasmodium genus, of which P. falciparum is the most prevalent. Several previous studies predicted human-plasmodium protein-protein interactions using computational methods have demonstrated their utility, accuracy, and efficiency to identify the interacting partners and therefore complementing experimental efforts to characterize host-pathogen interaction networks. To predict potential putative HP-PPIs, we use an integrative computational approach based on the combination of multiple OMICS-based methods including human red blood cells (RBC) and Plasmodium falciparum 3D7 strain expressed proteins, domain-domain based PPI, similarity of gene ontology terms, structure similarity method homology identification, and machine learning prediction. Our results reported a set of 716 protein interactions involving 302 human proteins and 130 Plasmodium proteins. This work provides a list of potential human-Plasmodium interacting proteins. These findings will contribute to better understand the mechanisms underlying the molecular determinism of malaria disease and potentially to identify candidate pharmacological targets.
41

Wilson, Jennifer L., Alessio Gravina, and Kevin Grimes. "From random to predictive: a context-specific interaction framework improves selection of drug protein–protein interactions for unknown drug pathways." Integrative Biology 14, no. 1 (January 2022): 13–24. http://dx.doi.org/10.1093/intbio/zyac002.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract With high drug attrition, protein–protein interaction (PPI) network models are attractive as efficient methods for predicting drug outcomes by analyzing proteins downstream of drug targets. Unfortunately, these methods tend to overpredict associations and they have low precision and prediction performance; performance is often no better than random (AUROC ~0.5). Typically, PPI models identify ranked phenotypes associated with downstream proteins, yet methods differ in prioritization of downstream proteins. Most methods apply global approaches for assessing all phenotypes. We hypothesized that a per-phenotype analysis could improve prediction performance. We compared two global approaches—statistical and distance-based—and our novel per-phenotype approach, ‘context-specific interaction’ (CSI) analysis, on severe side effect prediction. We used a novel dataset of adverse events (or designated medical events, DMEs) and discovered that CSI had a 50% improvement over global approaches (AUROC 0.77 compared to 0.51), and a 76–95% improvement in average precision (0.499 compared to 0.284, 0.256). Our results provide a quantitative rationale for considering downstream proteins on a per-phenotype basis when using PPI network methods to predict drug phenotypes.
42

Busler, Valerie J., Victor J. Torres, Mark S. McClain, Oscar Tirado, David B. Friedman, and Timothy L. Cover. "Protein-Protein Interactions among Helicobacter pylori Cag Proteins." Journal of Bacteriology 188, no. 13 (July 1, 2006): 4787–800. http://dx.doi.org/10.1128/jb.00066-06.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
ABSTRACT Many Helicobacter pylori isolates contain a 40-kb region of chromosomal DNA known as the cag pathogenicity island (PAI). The risk for development of gastric cancer or peptic ulcer disease is higher among humans infected with cag PAI-positive H. pylori strains than among those infected with cag PAI-negative strains. The cag PAI encodes a type IV secretion system that translocates CagA into gastric epithelial cells. To identify Cag proteins that are expressed by H. pylori during growth in vitro, we compared the proteomes of a wild-type H. pylori strain and an isogenic cag PAI deletion mutant using two-dimensional difference gel electrophoresis (2D-DIGE) in multiple pH ranges. Seven Cag proteins were identified by this approach. We then used a yeast two-hybrid system to detect potential protein-protein interactions among 14 Cag proteins. One heterotypic interaction (CagY/7 with CagX/8) and two homotypic interactions (involving H. pylori VirB11/ATPase and Cag5) were similar to interactions previously reported to occur among homologous components of the Agrobacterium tumefaciens type IV secretion system. Other interactions involved Cag proteins that do not have known homologues in other bacterial species. Biochemical analysis confirmed selected interactions involving five of the proteins that were identified by 2D-DIGE. Protein-protein interactions among Cag proteins are likely to have an important role in the assembly of the H. pylori type IV secretion apparatus.
43

Kosugi, Takatsugu, and Masahito Ohue. "Quantitative Estimate Index for Early-Stage Screening of Compounds Targeting Protein-Protein Interactions." International Journal of Molecular Sciences 22, no. 20 (October 10, 2021): 10925. http://dx.doi.org/10.3390/ijms222010925.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Drug-likeness quantification is useful for screening drug candidates. Quantitative estimates of drug-likeness (QED) are commonly used to assess quantitative drug efficacy but are not suitable for screening compounds targeting protein-protein interactions (PPIs), which have recently gained attention. Therefore, we developed a quantitative estimate index for compounds targeting PPIs (QEPPI), specifically for early-stage screening of PPI-targeting compounds. QEPPI is an extension of the QED method for PPI-targeting drugs that models physicochemical properties based on the information available for drugs/compounds, specifically those reported to act on PPIs. FDA-approved drugs and compounds in iPPI-DB, which comprise PPI inhibitors and stabilizers, were evaluated using QEPPI. The results showed that QEPPI is more suitable than QED for early screening of PPI-targeting compounds. QEPPI was also considered an extended concept of the “Rule-of-Four” (RO4), a PPI inhibitor index. We evaluated the discriminatory performance of QEPPI and RO4 for datasets of PPI-target compounds and FDA-approved drugs using F-score and other indices. The F-scores of RO4 and QEPPI were 0.451 and 0.501, respectively. QEPPI showed better performance and enabled quantification of drug-likeness for early-stage PPI drug discovery. Hence, it can be used as an initial filter to efficiently screen PPI-targeting compounds.
44

Sun, Xiao-Fei, and Salam Pradeep Singh. "Network pharmacology integrated molecular docking demonstrates the therapeutic mode of Panax ginseng against ovarian cancer." Tropical Journal of Pharmaceutical Research 22, no. 3 (April 19, 2023): 589–96. http://dx.doi.org/10.4314/tjpr.v22i3.16.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Purpose: To investigate the mode of action of the active ingredients of Panax ginseng against ovarian cancer protein targets elucidated using a network pharmacology approach and molecular docking study. Methods: An integrated protein-protein interactions (PPI) network targeting ovarian cancer and P. ginseng was constructed using a network pharmacology approach and molecular docking was carried out for the active constituents of P. ginseng against the target protein of P. ginseng. Results: Ninety-six compounds were used for the molecular docking simulation against five different proteins from a PPI network of proteins linked to ovarian cancer. The protein-ligand interaction showed strong molecular interaction at the active site of the proteins (p < 0.05).Conclusion: Ginsenosides and their derivatives present in P. ginseng successfully inhibit several key enzymes and proteins associated with ovarian cancer, which may act as potential therapeutic targets for the disease.
45

Goldsmith, Mark, Harto Saarinen, Guillermo García-Pérez, Joonas Malmi, Matteo A. C. Rossi, and Sabrina Maniscalco. "Link Prediction with Continuous-Time Classical and Quantum Walks." Entropy 25, no. 5 (April 28, 2023): 730. http://dx.doi.org/10.3390/e25050730.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein–protein interaction (PPI) networks consist of the physical and/or functional interactions between the proteins of an organism, and they form the basis for the field of network medicine. Since the biophysical and high-throughput methods used to form PPI networks are expensive, time-consuming, and often contain inaccuracies, the resulting networks are usually incomplete. In order to infer missing interactions in these networks, we propose a novel class of link prediction methods based on continuous-time classical and quantum walks. In the case of quantum walks, we examine the usage of both the network adjacency and Laplacian matrices for specifying the walk dynamics. We define a score function based on the corresponding transition probabilities and perform tests on six real-world PPI datasets. Our results show that continuous-time classical random walks and quantum walks using the network adjacency matrix can successfully predict missing protein–protein interactions, with performance rivalling the state-of-the-art.
46

Xu, Amy Y., Nicholas J. Clark, Joseph Pollastrini, Maribel Espinoza, Hyo-Jin Kim, Sekhar Kanapuram, Bruce Kerwin, et al. "Effects of Monovalent Salt on Protein-Protein Interactions of Dilute and Concentrated Monoclonal Antibody Formulations." Antibodies 11, no. 2 (March 31, 2022): 24. http://dx.doi.org/10.3390/antib11020024.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
In this study, we used sodium chloride (NaCl) to extensively modulate non-specific protein-protein interactions (PPI) of a humanized anti-streptavidin monoclonal antibody class 2 molecule (ASA-IgG2). The changes in PPI with varying NaCl (CNaCl) and monoclonal antibody (mAb) concentration (CmAb) were assessed using the diffusion interaction parameter kD and second virial coefficient B22 measured from solutions with low to moderate CmAb. The effective structure factor S(q)eff measured from concentrated mAb solutions using small-angle X-ray and neutron scattering (SAXS/SANS) was also used to characterize the PPI. Our results found that the nature of net PPI changed not only with CNaCl, but also with increasing CmAb. As a result, parameters measured from dilute and concentrated mAb samples could lead to different predictions on the stability of mAb formulations. We also compared experimentally determined viscosity results with those predicted from interaction parameters, including kD and S(q)eff. The lack of a clear correlation between interaction parameters and measured viscosity values indicates that the relationship between viscosity and PPI is concentration-dependent. Collectively, the behavior of flexible mAb molecules in concentrated solutions may not be correctly predicted using models where proteins are considered to be uniform colloid particles defined by parameters derived from low CmAb.
47

Li, Xue, Lifeng Yang, Xiaopan Zhang, and Xiong Jiao. "Prediction of Protein-Protein Interactions Based on Domain." Computational and Mathematical Methods in Medicine 2019 (August 21, 2019): 1–7. http://dx.doi.org/10.1155/2019/5238406.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Protein-protein interactions (PPIs) play a crucial role in various biological processes. To better comprehend the pathogenesis and treatments of various diseases, it is necessary to learn the detail of these interactions. However, the current experimental method still has many false-positive and false-negative problems. Computational prediction of protein-protein interaction has become a more important prediction method which can overcome the obstacles of the experimental method. In this work, we proposed a novel computational domain-based method for PPI prediction, and an SVM model for the prediction was built based on the physicochemical property of the domain. The outcomes of SVM and the domain-domain score were used to construct the prediction model for protein-protein interaction. The predicted results demonstrated the domain-based research can enhance the ability to predict protein interactions.
48

Li, Meijing, Tsendsuren Munkhdalai, Xiuming Yu, and Keun Ho Ryu. "A Novel Approach for Protein-Named Entity Recognition and Protein-Protein Interaction Extraction." Mathematical Problems in Engineering 2015 (2015): 1–10. http://dx.doi.org/10.1155/2015/942435.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Many researchers focus on developing protein-named entity recognition (Protein-NER) or PPI extraction systems. However, the studies about these two topics cannot be merged well; then existing PPI extraction systems’ Protein-NER still needs to improve. In this paper, we developed the protein-protein interaction extraction system named PPIMiner based on Support Vector Machine (SVM) and parsing tree. PPIMiner consists of three main models: natural language processing (NLP) model, Protein-NER model, and PPI discovery model. The Protein-NER model, which is named ProNER, identifies the protein names based on two methods: dictionary-based method and machine learning-based method. ProNER is capable of identifying more proteins than dictionary-based Protein-NER model in other existing systems. The final discovered PPIs extracted via PPI discovery model are represented in detail because we showed the protein interaction types and the occurrence frequency through two different methods. In the experiments, the result shows that the performances achieved by our ProNER and PPI discovery model are better than other existing tools. PPIMiner applied this protein-named entity recognition approach and parsing tree based PPI extraction method to improve the performance of PPI extraction. We also provide an easy-to-use interface to access PPIs database and an online system for PPIs extraction and Protein-NER.
49

Strotmann, Vivien I., and Yvonne Stahl. "Visualisation of in vivo protein-protein interactions." Journal of Experimental Botany, April 8, 2022. http://dx.doi.org/10.1093/jxb/erac139.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Molecular processes depend on the concerted and dynamic interactions of proteins, either by one-on-one interactions of the same or different proteins or by the assembly of larger protein complexes consisting of many different proteins. Here, not only the protein-protein interaction (PPI) itself, but also the localization and activity of the protein of interest (POI) within the cell is essential. Therefore, in all cell biological experiments, preserving the spatio-temporal state of one POI relative to another is key to understand the underlying complex and dynamic regulatory mechanisms in vivo. In this review we examine some of the applicable techniques to measure PPI in planta as well as recent combinatorial advances of PPI methods to measure the formation of higher order complexes with an emphasis on in vivo imaging techniques. We compare the different methods and discuss their benefits and potential pitfalls to facilitate the selection of appropriate techniques by giving a comprehensive overview about how to measure in vivo PPI in plants.
50

Wu, Zhourun, Qing Liao, Shixi Fan, and Bin Liu. "idenPC-CAP: Identify protein complexes from weighted RNA-protein heterogeneous interaction networks using co-assemble partner relation." Briefings in Bioinformatics, December 18, 2020. http://dx.doi.org/10.1093/bib/bbaa372.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Protein complexes play important roles in most cellular processes. The available genome-wide protein–protein interaction (PPI) data make it possible for computational methods identifying protein complexes from PPI networks. However, PPI datasets usually contain a large ratio of false positive noise. Moreover, different types of biomolecules in a living cell cooperate to form a union interaction network. Because previous computational methods focus only on PPIs ignoring other types of biomolecule interactions, their predicted protein complexes often contain many false positive proteins. In this study, we develop a novel computational method idenPC-CAP to identify protein complexes from the RNA-protein heterogeneous interaction network consisting of RNA–RNA interactions, RNA-protein interactions and PPIs. By considering interactions among proteins and RNAs, the new method reduces the ratio of false positive proteins in predicted protein complexes. The experimental results demonstrate that idenPC-CAP outperforms the other state-of-the-art methods in this field.

До бібліографії