1
|
Ilyas T, Ahmad K, Arsa DMS, Jeong YC, Kim H. Enhancing medical image analysis with unsupervised domain adaptation approach across microscopes and magnifications. Comput Biol Med 2024; 170:108055. [PMID: 38295480 DOI: 10.1016/j.compbiomed.2024.108055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/05/2024] [Accepted: 01/26/2024] [Indexed: 02/02/2024]
Abstract
In the domain of medical image analysis, deep learning models are heralding a revolution, especially in detecting complex and nuanced features characteristic of diseases like tumors and cancers. However, the robustness and adaptability of these models across varied imaging conditions and magnifications remain a formidable challenge. This paper introduces the Fourier Adaptive Recognition System (FARS), a pioneering model primarily engineered to address adaptability in malarial parasite recognition. Yet, the foundational principles guiding FARS lend themselves seamlessly to broader applications, including tumor and cancer diagnostics. FARS capitalizes on the untapped potential of transitioning from bounding box labels to richer semantic segmentation labels, enabling a more refined examination of microscopy slides. With the integration of adversarial training and the Color Domain Aware Fourier Domain Adaptation (F2DA), the model ensures consistent feature extraction across diverse microscopy configurations. The further inclusion of category-dependent context attention amplifies FARS's cross-domain versatility. Evidenced by a substantial elevation in cross-magnification performance from 31.3% mAP to 55.19% mAP and a 15.68% boost in cross-domain adaptability, FARS positions itself as a significant advancement in malarial parasite recognition. Furthermore, the core methodologies of FARS can serve as a blueprint for enhancing precision in other realms of medical image analysis, especially in the complex terrains of tumor and cancer imaging. The code is available at; https://github.com/Mr-TalhaIlyas/FARS.
Collapse
Affiliation(s)
- Talha Ilyas
- Division of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Core Research Institute of Intelligent Robots, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| | - Khubaib Ahmad
- Division of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Core Research Institute of Intelligent Robots, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Dewa Made Sri Arsa
- Division of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Core Research Institute of Intelligent Robots, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Department of Information Technology, Universitas Udayana, Bali, 80361, Indonesia
| | - Yong Chae Jeong
- Core Research Institute of Intelligent Robots, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Division of Electronics Engineering, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Hyongsuk Kim
- Division of Electronics and Information Engineering, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Core Research Institute of Intelligent Robots, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
2
|
Hayat M, Tahir M, Alarfaj FK, Alturki R, Gazzawe F. NLP-BCH-Ens: NLP-based intelligent computational model for discrimination of malaria parasite. Comput Biol Med 2022; 149:105962. [DOI: 10.1016/j.compbiomed.2022.105962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/29/2022] [Accepted: 08/13/2022] [Indexed: 11/03/2022]
|
3
|
Feng C, Wu J, Wei H, Xu L, Zou Q. CRCF: A Method of Identifying Secretory Proteins of Malaria Parasites. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2149-2157. [PMID: 34061749 DOI: 10.1109/tcbb.2021.3085589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Malaria is a mosquito-borne disease that results in millions of cases and deaths annually. The development of a fast computational method that identifies secretory proteins of the malaria parasite is important for research on antimalarial drugs and vaccines. Thus, a method was developed to identify the secretory proteins of malaria parasites. In this method, a reduced alphabet was selected to recode the original protein sequence. A feature synthesis method was used to synthesise three different types of feature information. Finally, the random forest method was used as a classifier to identify the secretory proteins. In addition, a web server was developed to share the proposed algorithm. Experiments using the benchmark dataset demonstrated that the overall accuracy achieved by the proposed method was greater than 97.8 percent using the 10-fold cross-validation method. Furthermore, the reduced schemes and characteristic performance analyses are discussed.
Collapse
|
4
|
Xia Z, Rong X, Dai Z, Zhou D. Identification of Novel Prognostic Biomarkers Relevant to Immune Infiltration in Lung Adenocarcinoma. Front Genet 2022; 13:863796. [PMID: 35571056 PMCID: PMC9092026 DOI: 10.3389/fgene.2022.863796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Programmed death ligand-1 (PD-L1) is a biomarker for assessing the immune microenvironment, prognosis, and response to immune checkpoint inhibitors in the clinical treatment of lung adenocarcinoma (LUAD), but it does not work for all patients. This study aims to discover alternative biomarkers. Methods: Public data were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Weighted gene co-expression network analysis (WGCNA) and gene ontology (GO) were used to determine the gene modules relevant to tumor immunity. Protein–protein interaction (PPI) network and GO semantic similarity analyses were applied to identify the module hub genes with functional similarities to PD-L1, and we assessed their correlations with immune infiltration, patient prognosis, and immunotherapy response. Immunohistochemistry (IHC) and hematoxylin and eosin (H&E) staining were used to validate the outcome at the protein level. Results: We identified an immune response–related module, and two hub genes (PSTPIP1 and PILRA) were selected as potential biomarkers with functional similarities to PD-L1. High expression levels of PSTPIP1 and PILRA were associated with longer overall survival and rich immune infiltration in LUAD patients, and both were significantly high in patients who responded to anti–PD-L1 treatment. Compared to PD-L1–negative LUAD tissues, the protein levels of PSTPIP1 and PILRA were relatively increased in the PD-L1–positive tissues, and the expression of PSTPIP1 and PILRA positively correlated with the tumor-infiltrating lymphocytes. Conclusion: We identified PSTPIP1 and PILRA as prognostic biomarkers relevant to immune infiltration in LUAD, and both are associated with the response to anti–PD-L1 treatment.
Collapse
Affiliation(s)
- Zhi Xia
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Xueyao Rong
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Ziyu Dai
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Dongbo Zhou
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Dongbo Zhou,
| |
Collapse
|
5
|
Xing J, Chen M, Han Y. Multiple datasets to explore the tumor microenvironment of cutaneous squamous cell carcinoma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:5905-5924. [PMID: 35603384 DOI: 10.3934/mbe.2022276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (cSCC) is one of the most frequent types of cutaneous cancer. The composition and heterogeneity of the tumor microenvironment significantly impact patient prognosis and the ability to practice precision therapy. However, no research has been conducted to examine the design of the tumor microenvironment and its interactions with cSCC. MATERIAL AND METHODS We retrieved the datasets GSE42677 and GSE45164 from the GEO public database, integrated them, and analyzed them using the SVA method. We then screened the core genes using the WGCNA network and LASSO regression and checked the model's stability using the ROC curve. Finally, we performed enrichment and correlation analyses on the core genes. RESULTS We identified four genes as core cSCC genes: DTYMK, CDCA8, PTTG1 and MAD2L1, and discovered that RORA, RORB and RORC were the primary regulators in the gene set. The GO semantic similarity analysis results indicated that CDCA8 and PTTG1 were the two most essential genes among the four core genes. The results of correlation analysis demonstrated that PTTG1 and HLA-DMA, CDCA8 and HLA-DQB2 were significantly correlated. CONCLUSIONS Examining the expression levels of four primary genes in cSCC aids in our understanding of the disease's pathophysiology. Additionally, the core genes were found to be highly related with immune regulatory genes, suggesting novel avenues for cSCC prevention and treatment.
Collapse
Affiliation(s)
- Jiahua Xing
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Muzi Chen
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
6
|
Liu Z, Li H, Pan S. Discovery and Validation of Key Biomarkers Based on Immune Infiltrates in Alzheimer's Disease. Front Genet 2021; 12:658323. [PMID: 34276768 PMCID: PMC8281057 DOI: 10.3389/fgene.2021.658323] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND As the most common neurodegenerative disease, Alzheimer's disease (AD) leads to progressive loss of cognition and memory. Presently, the underlying pathogenic genes of AD patients remain elusive, and effective disease-modifying therapy is not available. This study explored novel biomarkers that can affect diagnosis and treatment in AD based on immune infiltration. METHODS The gene expression profiles of 139 AD cases and 134 normal controls were obtained from the NCBI GEO public database. We applied the computational method CIBERSORT to bulk gene expression profiles of AD to quantify 22 subsets of immune cells. Besides, based on the use of the Least Absolute Shrinkage Selection Operator (LASSO), this study also applied SVM-RFE analysis to screen key genes. GO-based semantic similarity and logistic regression model analyses were applied to explore hub genes further. RESULTS There was a remarkable significance in the infiltration of immune cells between the subgroups. The proportions for monocytes, M0 macrophages, and dendritic cells in the AD group were significantly higher than those in the normal group, while the proportion of some cells was lower than that of the normal group, such as NK cell resting, T-cell CD4 naive, T-cell CD4 memory activation, and eosinophils. Additionally, seven genes (ABCA2, CREBRF, CD72, CETN2, KCNG1, NDUFA2, and RPL36AL) were identified as hub genes. Then we performed the analysis of immune factor correlation, gene set enrichment analysis (GSEA), and GO based on seven hub genes. The AUC of ROC prediction model in test and validation sets were 0.845 and 0.839, respectively. Eventually, the mRNA expression analysis of ABCA2, NDUFA2, CREBRF, and CD72 revealed significant differences among the seven hub genes and then was confirmed by RT-PCR. CONCLUSION A model based on immune cell infiltration might be used to forecast AD patients' diagnosis, and it provided a new perspective for AD treatment targets.
Collapse
Affiliation(s)
- Zhuohang Liu
- The Fifth Clinical Medical College of Anhui Medical University, Beijing, China
- Department of Hyperbaric Oxygen, Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hang Li
- Department of Hyperbaric Oxygen, Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuyi Pan
- The Fifth Clinical Medical College of Anhui Medical University, Beijing, China
- Department of Hyperbaric Oxygen, Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Moro G, Masseroli M. Gene function finding through cross-organism ensemble learning. BioData Min 2021; 14:14. [PMID: 33579334 PMCID: PMC7879670 DOI: 10.1186/s13040-021-00239-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 01/10/2021] [Indexed: 11/12/2022] Open
Abstract
Background Structured biological information about genes and proteins is a valuable resource to improve discovery and understanding of complex biological processes via machine learning algorithms. Gene Ontology (GO) controlled annotations describe, in a structured form, features and functions of genes and proteins of many organisms. However, such valuable annotations are not always reliable and sometimes are incomplete, especially for rarely studied organisms. Here, we present GeFF (Gene Function Finder), a novel cross-organism ensemble learning method able to reliably predict new GO annotations of a target organism from GO annotations of another source organism evolutionarily related and better studied. Results Using a supervised method, GeFF predicts unknown annotations from random perturbations of existing annotations. The perturbation consists in randomly deleting a fraction of known annotations in order to produce a reduced annotation set. The key idea is to train a supervised machine learning algorithm with the reduced annotation set to predict, namely to rebuild, the original annotations. The resulting prediction model, in addition to accurately rebuilding the original known annotations for an organism from their perturbed version, also effectively predicts new unknown annotations for the organism. Moreover, the prediction model is also able to discover new unknown annotations in different target organisms without retraining.We combined our novel method with different ensemble learning approaches and compared them to each other and to an equivalent single model technique. We tested the method with five different organisms using their GO annotations: Homo sapiens, Mus musculus, Bos taurus, Gallus gallus and Dictyostelium discoideum. The outcomes demonstrate the effectiveness of the cross-organism ensemble approach, which can be customized with a trade-off between the desired number of predicted new annotations and their precision.A Web application to browse both input annotations used and predicted ones, choosing the ensemble prediction method to use, is publicly available at http://tiny.cc/geff/. Conclusions Our novel cross-organism ensemble learning method provides reliable predicted novel gene annotations, i.e., functions, ranked according to an associated likelihood value. They are very valuable both to speed the annotation curation, focusing it on the prioritized new annotations predicted, and to complement known annotations available.
Collapse
Affiliation(s)
- Gianluca Moro
- DISI - University of Bologna, Via dell'Università, Cesena (FC), Italy.
| | - Marco Masseroli
- DEIB, Politecnico di Milano, Piazza L. Da Vinci 32, Milan, 20133, Italy
| |
Collapse
|
8
|
Domeniconi G, Masseroli M, Moro G, Pinoli P. Cross-organism learning method to discover new gene functionalities. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2016; 126:20-34. [PMID: 26724853 DOI: 10.1016/j.cmpb.2015.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 11/16/2015] [Accepted: 12/08/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Knowledge of gene and protein functions is paramount for the understanding of physiological and pathological biological processes, as well as in the development of new drugs and therapies. Analyses for biomedical knowledge discovery greatly benefit from the availability of gene and protein functional feature descriptions expressed through controlled terminologies and ontologies, i.e., of gene and protein biomedical controlled annotations. In the last years, several databases of such annotations have become available; yet, these valuable annotations are incomplete, include errors and only some of them represent highly reliable human curated information. Computational techniques able to reliably predict new gene or protein annotations with an associated likelihood value are thus paramount. METHODS Here, we propose a novel cross-organisms learning approach to reliably predict new functionalities for the genes of an organism based on the known controlled annotations of the genes of another, evolutionarily related and better studied, organism. We leverage a new representation of the annotation discovery problem and a random perturbation of the available controlled annotations to allow the application of supervised algorithms to predict with good accuracy unknown gene annotations. Taking advantage of the numerous gene annotations available for a well-studied organism, our cross-organisms learning method creates and trains better prediction models, which can then be applied to predict new gene annotations of a target organism. RESULTS We tested and compared our method with the equivalent single organism approach on different gene annotation datasets of five evolutionarily related organisms (Homo sapiens, Mus musculus, Bos taurus, Gallus gallus and Dictyostelium discoideum). Results show both the usefulness of the perturbation method of available annotations for better prediction model training and a great improvement of the cross-organism models with respect to the single-organism ones, without influence of the evolutionary distance between the considered organisms. The generated ranked lists of reliably predicted annotations, which describe novel gene functionalities and have an associated likelihood value, are very valuable both to complement available annotations, for better coverage in biomedical knowledge discovery analyses, and to quicken the annotation curation process, by focusing it on the prioritized novel annotations predicted.
Collapse
Affiliation(s)
- Giacomo Domeniconi
- DISI, Università degli Studi di Bologna, Via Venezia 52, 47521 Cesena, Italy.
| | - Marco Masseroli
- DEIB, Politecnico di Milano, Piazza L. Da Vinci 32, 20133 Milan, Italy.
| | - Gianluca Moro
- DISI, Università degli Studi di Bologna, Via Venezia 52, 47521 Cesena, Italy.
| | - Pietro Pinoli
- DEIB, Politecnico di Milano, Piazza L. Da Vinci 32, 20133 Milan, Italy.
| |
Collapse
|
9
|
Fan GL, Zhang XY, Liu YL, Nang Y, Wang H. DSPMP: Discriminating secretory proteins of malaria parasite by hybridizing different descriptors of Chou's pseudo amino acid patterns. J Comput Chem 2015; 36:2317-27. [PMID: 26484844 DOI: 10.1002/jcc.24210] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/20/2015] [Accepted: 08/23/2015] [Indexed: 12/28/2022]
Abstract
Identification of the proteins secreted by the malaria parasite is important for developing effective drugs and vaccines against infection. Therefore, we developed an improved predictor called "DSPMP" (Discriminating Secretory Proteins of Malaria Parasite) to identify the secretory proteins of the malaria parasite by integrating several vector features using support vector machine-based methods. DSPMP achieved an overall predictive accuracy of 98.61%, which is superior to that of the existing predictors in this field. We show that our method is capable of identifying the secretory proteins of the malaria parasite and found that the amino acid composition for buried and exposed sequences, denoted by AAC(b/e), was the most important feature for constructing the predictor. This article not only introduces a novel method for detecting the important features of sample proteins related to the malaria parasite but also provides a useful tool for tackling general protein-related problems. The DSPMP webserver is freely available at http://202.207.14.87:8032/fuwu/DSPMP/index.asp.
Collapse
Affiliation(s)
- Guo-Liang Fan
- Department of Physics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, 010021, China
| | - Xiao-Yan Zhang
- Department of Physics, Inner Mongolia University of Technology, Hohhot, 010051, China
| | - Yan-Ling Liu
- Department of Physics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, 010021, China
| | - Yi Nang
- Department of Physics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, 010021, China
| | - Hui Wang
- Department of Physics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, 010021, China
| |
Collapse
|
10
|
Feng YE. Identify Secretory Protein of Malaria Parasite with Modified Quadratic Discriminant Algorithm and Amino Acid Composition. Interdiscip Sci 2015; 8:156-161. [DOI: 10.1007/s12539-015-0112-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/15/2014] [Accepted: 03/16/2015] [Indexed: 12/13/2022]
|
11
|
Butler CL, Lucas O, Wuchty S, Xue B, Uversky VN, White M. Identifying novel cell cycle proteins in Apicomplexa parasites through co-expression decision analysis. PLoS One 2014; 9:e97625. [PMID: 24841368 PMCID: PMC4026381 DOI: 10.1371/journal.pone.0097625] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/22/2014] [Indexed: 11/26/2022] Open
Abstract
Hypothetical proteins comprise roughly half of the predicted gene complement of Toxoplasma gondii and Plasmodium falciparum and represent the largest class of uniquely functioning proteins in these parasites. Following the idea that functional relationships can be informed by the timing of gene expression, we devised a strategy to identify the core set of apicomplexan cell division cycling genes with important roles in parasite division, which includes many uncharacterized proteins. We assembled an expanded list of orthologs from the T. gondii and P. falciparum genome sequences (2781 putative orthologs), compared their mRNA profiles during synchronous replication, and sorted the resulting set of dual cell cycle regulated orthologs (744 total) into protein pairs conserved across many eukaryotic families versus those unique to the Apicomplexa. The analysis identified more than 100 ortholog gene pairs with unknown function in T. gondii and P. falciparum that displayed co-conserved mRNA abundance, dynamics of cyclical expression and similar peak timing that spanned the complete division cycle in each parasite. The unknown cyclical mRNAs encoded a diverse set of proteins with a wide range of mass and showed a remarkable conservation in the internal organization of ordered versus disordered structural domains. A representative sample of cyclical unknown genes (16 total) was epitope tagged in T. gondii tachyzoites yielding the discovery of new protein constituents of the parasite inner membrane complex, key mitotic structures and invasion organelles. These results demonstrate the utility of using gene expression timing and dynamic profile to identify proteins with unique roles in Apicomplexa biology.
Collapse
Affiliation(s)
- Carrie L. Butler
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Olivier Lucas
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Stefan Wuchty
- National Center for Biotechnology Information, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Bin Xue
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Michael White
- Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, United States of America
- Florida Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
12
|
Kuppuswamy U, Ananthasubramanian S, Wang Y, Balakrishnan N, Ganapathiraju MK. Predicting gene ontology annotations of orphan GWAS genes using protein-protein interactions. Algorithms Mol Biol 2014; 9:10. [PMID: 24708602 PMCID: PMC4124845 DOI: 10.1186/1748-7188-9-10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 03/11/2014] [Indexed: 01/30/2023] Open
Abstract
Background The number of genome-wide association studies (GWAS) has increased rapidly in the
past couple of years, resulting in the identification of genes associated with
different diseases. The next step in translating these findings into biomedically
useful information is to find out the mechanism of the action of these genes.
However, GWAS studies often implicate genes whose functions are currently unknown;
for example, MYEOV, ANKLE1, TMEM45B and ORAOV1 are found to be associated with
breast cancer, but their molecular function is unknown. Results We carried out Bayesian inference of Gene Ontology (GO) term annotations of genes
by employing the directed acyclic graph structure of GO and the network of
protein-protein interactions (PPIs). The approach is designed based on the fact
that two proteins that interact biophysically would be in physical proximity of
each other, would possess complementary molecular function, and play role in
related biological processes. Predicted GO terms were ranked according to their
relative association scores and the approach was evaluated quantitatively by
plotting the precision versus recall values and F-scores (the harmonic mean of
precision and recall) versus varying thresholds. Precisions of ~58%
and ~ 40% for localization and functions respectively of proteins were
determined at a threshold of ~30 (top 30 GO terms in the ranked list). Comparison
with function prediction based on semantic similarity among nodes in an ontology
and incorporation of those similarities in a k-nearest neighbor classifier
confirmed that our results compared favorably. Conclusions This approach was applied to predict the cellular component and molecular function
GO terms of all human proteins that have interacting partners possessing at least
one known GO annotation. The list of predictions is available at
http://severus.dbmi.pitt.edu/engo/GOPRED.html. We present the
algorithm, evaluations and the results of the computational predictions,
especially for genes identified in GWAS studies to be associated with diseases,
which are of translational interest.
Collapse
|
13
|
Predicting secretory proteins of malaria parasite by incorporating sequence evolution information into pseudo amino acid composition via grey system model. PLoS One 2012. [PMID: 23189138 PMCID: PMC3506597 DOI: 10.1371/journal.pone.0049040] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The malaria disease has become a cause of poverty and a major hindrance to economic development. The culprit of the disease is the parasite, which secretes an array of proteins within the host erythrocyte to facilitate its own survival. Accordingly, the secretory proteins of malaria parasite have become a logical target for drug design against malaria. Unfortunately, with the increasing resistance to the drugs thus developed, the situation has become more complicated. To cope with the drug resistance problem, one strategy is to timely identify the secreted proteins by malaria parasite, which can serve as potential drug targets. However, it is both expensive and time-consuming to identify the secretory proteins of malaria parasite by experiments alone. To expedite the process for developing effective drugs against malaria, a computational predictor called "iSMP-Grey" was developed that can be used to identify the secretory proteins of malaria parasite based on the protein sequence information alone. During the prediction process a protein sample was formulated with a 60D (dimensional) feature vector formed by incorporating the sequence evolution information into the general form of PseAAC (pseudo amino acid composition) via a grey system model, which is particularly useful for solving complicated problems that are lack of sufficient information or need to process uncertain information. It was observed by the jackknife test that iSMP-Grey achieved an overall success rate of 94.8%, remarkably higher than those by the existing predictors in this area. As a user-friendly web-server, iSMP-Grey is freely accessible to the public at http://www.jci-bioinfo.cn/iSMP-Grey. Moreover, for the convenience of most experimental scientists, a step-by-step guide is provided on how to use the web-server to get the desired results without the need to follow the complicated mathematical equations involved in this paper.
Collapse
|
14
|
Han Y, Yu G, Sarioglu H, Caballero-Martinez A, Schlott F, Ueffing M, Haase H, Peschel C, Krackhardt AM. Proteomic investigation of the interactome of FMNL1 in hematopoietic cells unveils a role in calcium-dependent membrane plasticity. J Proteomics 2012. [PMID: 23182705 DOI: 10.1016/j.jprot.2012.11.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Formin-like 1 (FMNL1) is a formin-related protein highly expressed in hematopoietic cells and overexpressed in leukemias as well as diverse transformed cell lines. It has been described to play a role in diverse functions of hematopoietic cells such as phagocytosis of macrophages as well as polarization and cytotoxicity of T cells. However, the specific role of FMNL1 in these processes has not been clarified yet and regulation by interaction partners in primary hematopoietic cells has never been investigated. We performed a proteomic screen for investigation of the interactome of FMNL1 in primary hematopoietic cells resulting in the identification of a number of interaction partners. Bioinformatic analysis considering semantic similarity suggested the giant protein AHNAK1 to be an essential interaction partner of FMNL1. We confirmed AHNAK1 as a general binding partner for FMNL1 in diverse hematopoietic cells and demonstrate that the N-terminal part of FMNL1 binds to the C-terminus of AHNAK1. Moreover, we show that the constitutively activated form of FMNL1 (FMNL1γ) induces localization of AHNAK1 to the cell membrane. Finally, we provide evidence that overexpression or knock down of FMNL1 has an impact on the capacitative calcium influx after ionomycin-mediated activation of diverse cell lines and primary cells.
Collapse
Affiliation(s)
- Yanan Han
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Wang J, Zhou X, Zhu J, Gu Y, Zhao W, Zou J, Guo Z. GO-function: deriving biologically relevant functions from statistically significant functions. Brief Bioinform 2011; 13:216-27. [DOI: 10.1093/bib/bbr041] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|