Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Curr Issues Mol Biol ; 45(11): 8652-8669, 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37998721

ABSTRACT

Advancements in molecular biology have revolutionized our understanding of complex diseases, with Alzheimer's disease being a prime example. Single-cell sequencing, currently the most suitable technology, facilitates profoundly detailed disease analysis at the cellular level. Prior research has established that the pathology of Alzheimer's disease varies across different brain regions and cell types. In parallel, only machine learning has the capacity to address the myriad challenges presented by such studies, where the integration of large-scale data and numerous experiments is required to extract meaningful knowledge. Our methodology utilizes single-cell RNA sequencing data from healthy and Alzheimer's disease (AD) samples, focused on the cortex and hippocampus regions in mice. We designed three distinct case studies and implemented an ensemble feature selection approach through machine learning, also performing an analysis of distinct age-related datasets to unravel age-specific effects, showing differential gene expression patterns within each condition. Important evidence was reported, such as enrichment in central nervous system development and regulation of oligodendrocyte differentiation between the hippocampus and cortex of 6-month-old AD mice as well as regulation of epinephrine secretion and dendritic spine morphogenesis in 15-month-old AD mice. Our outcomes from all three of our case studies illustrate the capacity of machine learning strategies when applied to single-cell data, revealing critical insights into Alzheimer's disease.

2.
Adv Exp Med Biol ; 1424: 241-246, 2023.
Article in English | MEDLINE | ID: mdl-37486500

ABSTRACT

The high-throughput sequencing method known as RNA-Seq records the whole transcriptome of individual cells. Single-cell RNA sequencing, also known as scRNA-Seq, is widely utilized in the field of biomedical research and has resulted in the generation of huge quantities and types of data. The noise and artifacts that are present in the raw data require extensive cleaning before they can be used. When applied to applications for machine learning or pattern recognition, feature selection methods offer a method to reduce the amount of time spent on calculation while simultaneously improving predictions and offering a better knowledge of the data. The process of discovering biomarkers is analogous to feature selection methods used in machine learning and is especially helpful for applications in the medical field. An attempt is made by a feature selection algorithm to cut down on the total number of features by eliminating those that are unnecessary or redundant while retaining those that are the most helpful.We apply FS algorithms designed for scRNA-Seq to Alzheimer's disease, which is the most prevalent neurodegenerative disease in the western world and causes cognitive and behavioral impairment. AD is clinically and pathologically varied, and genetic studies imply a diversity of biological mechanisms and pathways. Over 20 new Alzheimer's disease susceptibility loci have been discovered through linkage, genome-wide association, and next-generation sequencing (Tosto G, Reitz C, Mol Cell Probes 30:397-403, 2016). In this study, we focus on the performance of three different approaches to marker gene selection methods and compare them using the support vector machine (SVM), k-nearest neighbors' algorithm (k-NN), and linear discriminant analysis (LDA), which are mainly supervised classification algorithms.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Humans , Alzheimer Disease/genetics , Genome-Wide Association Study , Algorithms , RNA-Seq
3.
Adv Exp Med Biol ; 1423: 201-206, 2023.
Article in English | MEDLINE | ID: mdl-37525045

ABSTRACT

Protein folding is the process by which a polypeptide chain self-assembles into the correct three-dimensional structure, so that it ends up in the biologically active, native state. Under conditions of proteotoxic stress, mutations, or cellular aging, proteins can begin to aggregate into non-native structures such as ordered amyloid fibrils and plaques. Many neurodegenerative diseases involve the misfolding and aggregation of specific proteins into abnormal, toxic species. Experimental approaches including crystallography and AFM (atomic force microscopy)-based force spectroscopy are used to exploit the folding and structural characterization of protein molecules. At the same time, computational techniques through molecular dynamics, fold recognition, and structure prediction are widely applied in this direction. Benchmarking analysis for combining and comparing computational methodologies with functional studies can decisively unravel robust interactions between the side groups of the amino acid sequence and monitor alterations in intrinsic protein dynamics with high precision as well as adequately determine potent conformations of the folded patterns formed in the polypeptide structure.


Subject(s)
Peptides , Protein Folding , Amino Acid Sequence , Amyloid/chemistry , Molecular Dynamics Simulation , Protein Conformation
4.
Adv Exp Med Biol ; 1423: 207-214, 2023.
Article in English | MEDLINE | ID: mdl-37525046

ABSTRACT

System-level network-based approaches are an emerging field in the biomedical domain since biological networks can be used to analyze complicated biological processes and complex human disorders more efficiently. Network biomarkers are groups of interconnected molecular components causing perturbations in the entire network topology that can be used as indicators of pathogenic biological processes when studying a given disease. Although in the last years computational systems-based approaches have gained ground on the path to discovering new network biomarkers, in complex diseases like Alzheimer's disease (AD), this approach has still much to offer. Especially the adoption of single-cell RNA sequencing (scRNA-seq) has now become the dominant technology for the study of stochastic gene expression. Toward this orientation, we propose an R workflow that extracts disease-perturbed subpathways within a pathway network. We construct a gene-gene interaction network integrated with scRNA-seq expression profiles, and after network processing and pruning, the most active subnetworks are isolated from the entire network topology. The proposed methodology was applied on a real AD-based scRNA-seq data, providing already existing and new potential AD biomarkers in gene network context.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Biomarkers , Gene Regulatory Networks , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
5.
Adv Exp Med Biol ; 1423: 215-224, 2023.
Article in English | MEDLINE | ID: mdl-37525047

ABSTRACT

Gene regulatory network (GRN) inference from gene expression data is a highly complex and challenging task in systems biology. Despite the challenges, GRNs have emerged, and for complex diseases such as neurodegenerative diseases, they have the potential to provide vital information and identify key regulators. However, every GRN method produced predicts results based on its assumptions, providing limited biological insights. For that reason, the current work focused on the development of an ensemble method from individual GRN methods to address this issue. Four state-of-the-art GRN algorithms were selected to form a consensus GRN from their common gene interactions. Each algorithm uses a different construction method, and for a more robust behavior, both static and dynamic methods were selected as well. The algorithms were applied to a scRNA-seq dataset from the CK-p25 mus musculus model during neurodegeneration. The top subnetworks were constructed from the consensus network, and potential key regulators were identified. The results also demonstrated the overlap between the algorithms for the current dataset and the necessity for an ensemble approach. This work aims to demonstrate the creation of an ensemble network and provide insights into whether a combination of different GRN methods can produce valuable results.


Subject(s)
Gene Regulatory Networks , Neurodegenerative Diseases , Animals , Mice , Humans , Neurodegenerative Diseases/genetics , Consensus , Single-Cell Gene Expression Analysis , Computational Biology/methods , Algorithms
6.
Adv Exp Med Biol ; 1424: 23-29, 2023.
Article in English | MEDLINE | ID: mdl-37486475

ABSTRACT

Biosensing platforms have gained much attention in clinical practice screening thousands of samples simultaneously for the accurate detection of important markers in various diseases for diagnostic and prognostic purposes. Herein, a framework for the design of an innovative methodological approach combined with data processing and appropriate software in order to implement a complete diagnostic system for Parkinson's disease exploitation is presented. The integrated platform consists of biochemical and peripheral sensor platforms for measuring biological and biometric parameters of examinees, a central collection and management unit along with a server for storing data, and a decision support system for patient's state assessment regarding the occurrence of the disease. The suggested perspective is oriented on data processing and experimental implementation and can provide a powerful holistic evaluation of personalized monitoring of patients or individuals at high risk of manifestation of the disease.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/diagnosis , Software
7.
Adv Exp Med Biol ; 1423: 1-10, 2023.
Article in English | MEDLINE | ID: mdl-37525028

ABSTRACT

The clinical pathology of neurodegenerative diseases suggests that earlier onset and progression are related to the accumulation of protein aggregates due to misfolding. A prominent way to extract useful information regarding single-molecule studies of protein misfolding at the nanoscale is by capturing the unbinding molecular forces through forced mechanical tension generated and monitored by an atomic force microscopy-based single-molecule force spectroscopy (AFM-SMFS). This AFM-driven process results in an amount of data in the form of force versus molecular extension plots (force-distance curves), the statistical analysis of which can provide insights into the underlying energy landscape and assess a number of characteristic elastic and kinetic molecular parameters of the investigated sample. This chapter outlines the setup of a bio-AFM-based SMFS technique for single-molecule probing. The infrastructure used as a reference for this presentation is the Bruker ForceRobot300.


Subject(s)
Neurodegenerative Diseases , Humans , Proteins/chemistry , Microscopy, Atomic Force/methods , Nanotechnology , Single Molecule Imaging
8.
Adv Exp Med Biol ; 1424: 187-192, 2023.
Article in English | MEDLINE | ID: mdl-37486493

ABSTRACT

The increase in the population's life expectancy leads to an increase in the incidence of dementia and, therefore, in diseases such as Alzheimer's. Towards this direction, the HELIAD1 study is the first large-scale epidemiological study aimed at assessing epidemiological data on dementia, mild mental decline, and other neuropsychiatric disorders associated with old age. This is a huge study with several computational challenges, most of which can be addressed by machine learning processes. The objectives of this study were to detect patterns in the HELIAD clinical data that classify with high accuracy various levels of cognitive impairment by training ML algorithms and hence apply derived model on future clinical data to predict with the same accuracy the class variable. We propose a machine learning method based on RUSBoost classifier to identify a critical subset of biomarkers that classify accurately between neurological patients with mild cognitive impairment (MCI) or dementia of the Alzheimer's type (DAT) and the cognitively healthy control (CHC) group. In this study we used a highly skewed (imbalanced) dataset with most observations (majority class) belonging to the CHC group. The method proposed predicts accurately the clinical diagnosis label and effectively classifies the neurological patients from the CHC class. In particular, the classification accuracy (actual vs predicted) for the three classes of the clinical diagnosis was 97%, 78%, and 91% for control, MCI, and dementia class, respectively.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/epidemiology , Alzheimer Disease/complications , Sensitivity and Specificity , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/complications , Machine Learning , Biomarkers , Disease Progression
9.
Sensors (Basel) ; 23(9)2023 Apr 22.
Article in English | MEDLINE | ID: mdl-37177386

ABSTRACT

Alzheimer's disease (AD) is now classified as a silent pandemic due to concerning current statistics and future predictions. Despite this, no effective treatment or accurate diagnosis currently exists. The negative impacts of invasive techniques and the failure of clinical trials have prompted a shift in research towards non-invasive treatments. In light of this, there is a growing need for early detection of AD through non-invasive approaches. The abundance of data generated by non-invasive techniques such as blood component monitoring, imaging, wearable sensors, and bio-sensors not only offers a platform for more accurate and reliable bio-marker developments but also significantly reduces patient pain, psychological impact, risk of complications, and cost. Nevertheless, there are challenges concerning the computational analysis of the large quantities of data generated, which can provide crucial information for the early diagnosis of AD. Hence, the integration of artificial intelligence and deep learning is critical to addressing these challenges. This work attempts to examine some of the facts and the current situation of these approaches to AD diagnosis by leveraging the potential of these tools and utilizing the vast amount of non-invasive data in order to revolutionize the early detection of AD according to the principles of a new non-invasive medicine era.


Subject(s)
Alzheimer Disease , Deep Learning , Humans , Artificial Intelligence , Alzheimer Disease/diagnosis , Biomarkers , Early Diagnosis
10.
Sensors (Basel) ; 22(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35062370

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with dysfunction of dopaminergic neurons in the brain, lack of dopamine and the formation of abnormal Lewy body protein particles. PD is an idiopathic disease of the nervous system, characterized by motor and nonmotor manifestations without a discrete onset of symptoms until a substantial loss of neurons has already occurred, enabling early diagnosis very challenging. Sensor-based platforms have gained much attention in clinical practice screening various biological signals simultaneously and allowing researchers to quickly receive a huge number of biomarkers for diagnostic and prognostic purposes. The integration of machine learning into medical systems provides the potential for optimization of data collection, disease prediction through classification of symptoms and can strongly support data-driven clinical decisions. This work attempts to examine some of the facts and current situation of sensor-based approaches in PD diagnosis and discusses ensemble techniques using sensor-based data for developing machine learning models for personalized risk prediction. Additionally, a biosensing platform combined with clinical data processing and appropriate software is proposed in order to implement a complete diagnostic system for PD monitoring.


Subject(s)
Parkinson Disease , Brain , Dopamine , Dopaminergic Neurons , Humans , Machine Learning , Parkinson Disease/diagnosis
11.
Adv Exp Med Biol ; 1338: 135-144, 2021.
Article in English | MEDLINE | ID: mdl-34973018

ABSTRACT

In the last two decades, the medical sciences have changed their approach to pathogenesis as well as to the diagnosis and treatment of complex human diseases. The main reason for this change is the explosive development of biomedical technology and research, which produces a huge amount of information and data which are generated at an increasing rate. Toward this direction is the pathway analysis, a thriving research area of systems biology tools and methodologies which aim to unravel the inherent complexity of high-throughput biological data produced by the advent of omics technologies. Through this graph mining approach, we can deal with the complexity of the cellular systems of various diseases such as Alzheimer's disease. In this work, we developed a subpathway analysis method for single-cell RNA-seq experiments which isolates differentially expressed subpathways indicating potentially perturbed biological processes. The differential expression status of each gene is negotiated among well-established RNA-seq differential expression analysis tools in order to minimize false discoveries. Also, we demonstrate the efficacy of our method on a single-cell RNA-seq dataset for temporal tracking of microglia activation in neurodegeneration. Results suggest that our approach succeeds in isolating several perturbed biological processes known to be associated with neurodegeneration.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Humans , Systems Biology
12.
Adv Exp Med Biol ; 1338: 199-208, 2021.
Article in English | MEDLINE | ID: mdl-34973026

ABSTRACT

We live in the big data era in the biomedical field, where machine learning has a very important contribution to the interpretation of complex biological processes and diseases, since it has the potential to create predictive models from multidimensional data sets. Part of the application of machine learning in biomedical science is to study and model complex cellular systems such as biological networks. In this context, the study of complex diseases, such as Alzheimer's diseases (AD), benefits from established methodologies of network science and machine learning as they offer algorithmic tools and techniques that can address the limitations and challenges of modeling and studying cellular AD-related networks. In this paper we analyze the opportunities and challenges at the intersection of machine learning and network biology and whether this can affect the biological interpretation and clarification of diseases. Specifically, we focus on GRN techniques which through omics data and the use of machine learning techniques can construct a network that captures all the information at the molecular level for the disease under study. We record the emerging machine learning techniques that are focus on ensemble tree-based techniques in the area of classification and regression. Their potential for unraveling the complexity of model cellular systems in complex diseases, such as AD, offers the opportunity for novel machine learning methodologies to decipher the mechanisms of the various AD processes.


Subject(s)
Alzheimer Disease , Humans , Machine Learning , Models, Biological
13.
Adv Exp Med Biol ; 1194: 303-314, 2020.
Article in English | MEDLINE | ID: mdl-32468546

ABSTRACT

MOTIVATION: In the last years, systems-level network-based approaches have gained ground in the research field of systems biology. These approaches are based on the analysis of high-throughput sequencing studies, which are rapidly increasing year by year. Nowadays, the single-cell RNA-sequencing, an optimized next-generation sequencing (NGS) technology that offers a better understanding of the function of an individual cell in the context of its microenvironment, prevails. RESULTS: Toward this direction, a method is developed in which active molecular subpathways are recorded during the time evolution of the disease under study. This method operates for expression profiling by high-throughput sequencing data. Its capability is based on capturing the temporal changes of local gene communities that form a disease-perturbed subpathway. The aforementioned methods are applied to real data from a recent study that uses single-cell RNA-sequencing data related with the progression of neurodegeneration. More specific, microglia cells were isolated from the hippocampus of a mouse model with Alzheimer's disease-like phenotypes and severe neurodegeneration and of control mice at multiple time points during progression of neurodegeneration. Our analysis offers a different view for neurodegeneration progression under the perspective of systems biology. CONCLUSION: Our approach into the molecular perspective using a temporal tracking of active pathways in neurodegeneration at single-cell resolution may offer new insights for designing new efficient strategies to treat Alzheimer's and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Systems Biology , Alzheimer Disease/physiopathology , Animals , Disease Models, Animal , Disease Progression , Humans , Mice , Microglia/pathology , Sequence Analysis, RNA , Single-Cell Analysis/standards , Systems Biology/methods
14.
Adv Exp Med Biol ; 1194: 409-421, 2020.
Article in English | MEDLINE | ID: mdl-32468556

ABSTRACT

MotivationNeurodegenerative diseases (NDs), including amyotrophic lateral sclerosis, Parkinson's disease, Alzheimer's disease, and Huntington's disease, occur as a result of neurodegenerative processes. Thus, it has been increasingly appreciated that many neurodegenerative conditions overlap at multiple levels. However, traditional clinicopathological correlation approaches to better classify a disease have met with limited success. Discovering this overlap offers hope for therapeutic advances that could ameliorate many ND simultaneously. In parallel, in the last decade, systems biology approaches have become a reliable choice in complex disease analysis for gaining more delicate biological insights and have enabled the comprehension of the higher order functions of the biological systems.ResultsToward this orientation, we developed a systems biology approach for the identification of common links and pathways of ND, based on well-established and novel topological and functional measures. For this purpose, a molecular pathway network was constructed, using molecular interactions and relations of four main neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease). Our analysis captured the overlapped subregions forming molecular subpathways fully enriched in these four NDs. Also, it exported molecules that act as bridges, hubs, and key players for neurodegeneration concerning either their topology or their functional role.ConclusionUnderstanding these common links and central topologies under the perspective of systems biology and network theory and greater insights are provided to uncover the complex neurodegeneration processes.


Subject(s)
Neurodegenerative Diseases , Systems Biology , Humans , Neural Pathways/pathology , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/physiopathology
15.
Bioinformatics ; 32(24): 3844-3846, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27542770

ABSTRACT

DEsubs is a network-based systems biology R package that extracts disease-perturbed subpathways within a pathway network as recorded by RNA-seq experiments. It contains an extensive and customized framework with a broad range of operation modes at all stages of the subpathway analysis, enabling so a case-specific approach. The operation modes include pathway network construction and processing, subpathway extraction, visualization and enrichment analysis with regard to various biological and pharmacological features. Its capabilities render DEsubs a tool-guide for both the modeler and experimentalist for the identification of more robust systems-level drug targets and biomarkers for complex diseases. AVAILABILITY AND IMPLEMENTATION: DEsubs is implemented as an R package following Bioconductor guidelines: http://bioconductor.org/packages/DEsubs/ CONTACT: tassos.bezerianos@nus.edu.sgSupplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Sequence Analysis, RNA/methods , Software , Humans , RNA , Transcriptome
16.
Bioinformatics ; 32(6): 884-92, 2016 03 15.
Article in English | MEDLINE | ID: mdl-26568631

ABSTRACT

MOTIVATION: In the era of network medicine and the rapid growth of paired time series mRNA/microRNA expression experiments, there is an urgent need for pathway enrichment analysis methods able to capture the time- and condition-specific 'active parts' of the biological circuitry as well as the microRNA impact. Current methods ignore the multiple dynamical 'themes'-in the form of enriched biologically relevant microRNA-mediated subpathways-that determine the functionality of signaling networks across time. RESULTS: To address these challenges, we developed time-vaRying enriCHment integrOmics Subpathway aNalysis tOol (CHRONOS) by integrating time series mRNA/microRNA expression data with KEGG pathway maps and microRNA-target interactions. Specifically, microRNA-mediated subpathway topologies are extracted and evaluated based on the temporal transition and the fold change activity of the linked genes/microRNAs. Further, we provide measures that capture the structural and functional features of subpathways in relation to the complete organism pathway atlas. Our application to synthetic and real data shows that CHRONOS outperforms current subpathway-based methods into unraveling the inherent dynamic properties of pathways. AVAILABILITY AND IMPLEMENTATION: CHRONOS is freely available at http://biosignal.med.upatras.gr/chronos/ CONTACT: tassos.bezerianos@nus.edu.sg SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
MicroRNAs/genetics , Signal Transduction
17.
Adv Exp Med Biol ; 988: 215-224, 2017.
Article in English | MEDLINE | ID: mdl-28971401

ABSTRACT

In the era of Systems Biology and growing flow of omics experimental data from high throughput techniques, experimentalists are in need of more precise pathway-based tools to unravel the inherent complexity of diseases and biological processes. Subpathway-based approaches are the emerging generation of pathway-based analysis elucidating the biological mechanisms under the perspective of local topologies onto a complex pathway network. Towards this orientation, we developed PerSub, a graph-based algorithm which detects subpathways perturbed by a complex disease. The perturbations are imprinted through differentially expressed and co-expressed subpathways as recorded by RNA-seq experiments. Our novel algorithm is applied on data obtained from a real experimental study and the identified subpathways provide biological evidence for the brain aging.


Subject(s)
Algorithms , Systems Biology , RNA
18.
BMC Genomics ; 16: 147, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25887273

ABSTRACT

BACKGROUND: The avalanche of integromics and panomics approaches shifted the deciphering of aging mechanisms from single molecular entities to communities of them. In this orientation, we explore the cardiac aging mechanisms - risk factor for multiple cardiovascular diseases - by capturing the micronome synergism and detecting longevity signatures in the form of communities (modules). For this, we developed a meta-analysis scheme that integrates transcriptome expression data from multiple cardiac-specific independent studies in mouse and human along with proteome and micronome interaction data in the form of multiple independent weighted networks. Modularization of each weighted network produced modules, which in turn were further analyzed so as to define consensus modules across datasets that change substantially during lifespan. Also, we established a metric that determines - from the modular perspective - the synergism of microRNA-microRNA interactions as defined by significantly functionally associated targets. RESULTS: The meta-analysis provided 40 consensus integromics modules across mouse datasets and revealed microRNA relations with substantial collective action during aging. Three modules were reproducible, based on homology, when mapped against human-derived modules. The respective homologs mainly represent NADH dehydrogenases, ATP synthases, cytochrome oxidases, Ras GTPases and ribosomal proteins. Among various observations, we corroborate to the involvement of miR-34a (included in consensus modules) as proposed recently; yet we report that has no synergistic effect. Moving forward, we determined its age-related neighborhood in which HCN3, a known heart pacemaker channel, was included. Also, miR-125a-5p/-351, miR-200c/-429, miR-106b/-17, miR-363/-92b, miR-181b/-181d, miR-19a/-19b, let-7d/-7f, miR-18a/-18b, miR-128/-27b and miR-106a/-291a-3p pairs exhibited significant synergy and their association to aging and/or cardiovascular diseases is supported in many cases by a disease database and previous studies. On the contrary, we suggest that miR-22 has not substantial impact on heart longevity as proposed recently. CONCLUSIONS: We revised several proteins and microRNAs recently implicated in cardiac aging and proposed for the first time modules as signatures. The integromics meta-analysis approach can serve as an efficient subvening signature tool for more-oriented better-designed experiments. It can also promote the combinational multi-target microRNA therapy of age-related cardiovascular diseases along the continuum from prevention to detection, diagnosis, treatment and outcome.


Subject(s)
Aging/genetics , Cardiovascular Diseases/genetics , MicroRNAs/genetics , Transcriptome/genetics , Aging/pathology , Animals , Cardiovascular Diseases/pathology , Gene Regulatory Networks , Heart/physiopathology , Humans , Mice
19.
Article in English | MEDLINE | ID: mdl-36767399

ABSTRACT

Since December 2019, the coronavirus disease has significantly affected millions of people. Given the effect this disease has on the pulmonary systems of humans, there is a need for chest radiographic imaging (CXR) for monitoring the disease and preventing further deaths. Several studies have been shown that Deep Learning models can achieve promising results for COVID-19 diagnosis towards the CXR perspective. In this study, five deep learning models were analyzed and evaluated with the aim of identifying COVID-19 from chest X-ray images. The scope of this study is to highlight the significance and potential of individual deep learning models in COVID-19 CXR images. More specifically, we utilized the ResNet50, ResNet101, DenseNet121, DenseNet169 and InceptionV3 using Transfer Learning. All models were trained and validated on the largest publicly available repository for COVID-19 CXR images. Furthermore, they were evaluated on unknown data that was not used for training or validation, authenticating their performance and clarifying their usage in a medical scenario. All models achieved satisfactory performance where ResNet101 was the superior model achieving 96% in Precision, Recall and Accuracy, respectively. Our outcomes show the potential of deep learning models on COVID-19 medical offering a promising way for the deeper understanding of COVID-19.


Subject(s)
COVID-19 , Deep Learning , Humans , COVID-19/diagnostic imaging , COVID-19 Testing , X-Rays , Thorax
20.
J Bioinform Comput Biol ; 21(5): 2340002, 2023 10.
Article in English | MEDLINE | ID: mdl-37743364

ABSTRACT

The evolution of single-cell technology is ongoing, continually generating massive amounts of data that reveal many mysteries surrounding intricate diseases. However, their drawbacks continue to constrain us. Among these, annotating cell types in single-cell gene expressions pose a substantial challenge, despite the myriad of tools at our disposal. The rapid growth in data, resources, and tools has consequently brought about significant alterations in this area over the years. In our study, we spotlight all note-worthy cell type annotation techniques developed over the past four years. We provide an overview of the latest trends in this field, showcasing the most advanced methods in taxonomy. Our research underscores the demand for additional tools that incorporate a biological context and also predicts that the rising trend of graph neural network approaches will likely lead this research field in the coming years.


Subject(s)
Neural Networks, Computer , Single-Cell Gene Expression Analysis , Sequence Analysis, RNA , Gene Expression Profiling
SELECTION OF CITATIONS
SEARCH DETAIL