1
|
Ren J, Gao Q, Zhou X, Chen L, Guo W, Feng K, Hu J, Huang T, Cai YD. Identification of gene and protein signatures associated with long-term effects of COVID-19 on the immune system after patient recovery by analyzing single-cell multi-omics data using a machine learning approach. Vaccine 2024; 42:126253. [PMID: 39182316 DOI: 10.1016/j.vaccine.2024.126253] [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: 01/08/2024] [Revised: 08/17/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
Viral infections significantly impact the immune system, and impact will persist until recovery. However, the influence of severe acute respiratory syndrome coronavirus 2 infection on the homeostatic immune status and secondary immune response in recovered patients remains unclear. To investigate these persistent alterations, we employed five feature-ranking algorithms (LASSO, MCFS, RF, CATBoost, and XGBoost), incremental feature selection, synthetic minority oversampling technique and two classification algorithms (decision tree and k-nearest neighbors) to analyze multi-omics data (surface proteins and transcriptome) from coronavirus disease 2019 (COVID-19) recovered patients and healthy controls post-influenza vaccination. The single-cell multi-omics dataset was divided into five subsets corresponding to five immune cell subtypes: B cells, CD4+ T cells, CD8+ T cells, Monocytes, and Natural Killer cells. Each cell was represented by 28,402 scRNA-seq (RNA) features, 3 Hash Tag Oligo (HTO) features, 138 Cellular indexing of transcriptomes and epitopes by sequencing (CITE) features and 23,569 Single Cell Transform (SCT) features. Some multi-omics markers were identified and effective classifiers were constructed. Our findings indicate a distinct immune status in COVID-19 recovered patients, characterized by low expression of ribosomal protein (RPS26) and high expression of immune cell surface proteins (CD33, CD48). Notably, TMEM176B, a membrane protein, was highly expressed in monocytes of COVID-19 convalescent patients. These observations aid in discerning molecular differences among immune cell subtypes and contribute to understanding the prolonged effects of COVID-19 on the immune system, which is valuable for treating infectious diseases like COVID-19.
Collapse
Affiliation(s)
- JingXin Ren
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Qian Gao
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - XianChao Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China
| | - Jerry Hu
- Department of Natural Sciences and Mathematics, College of Natural and Applied Science, University of Houston - Victoria, Victoria, TX 77901, USA.
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
2
|
Zheng Y, Schupp JC, Adams T, Clair G, Justet A, Ahangari F, Yan X, Hansen P, Carlon M, Cortesi E, Vermant M, Vos R, De Sadeleer LJ, Rosas IO, Pineda R, Sembrat J, Königshoff M, McDonough JE, Vanaudenaerde BM, Wuyts WA, Kaminski N, Ding J. Unagi: Deep Generative Model for Deciphering Cellular Dynamics and In-Silico Drug Discovery in Complex Diseases. RESEARCH SQUARE 2023:rs.3.rs-3676579. [PMID: 38196613 PMCID: PMC10775382 DOI: 10.21203/rs.3.rs-3676579/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Human diseases are characterized by intricate cellular dynamics. Single-cell sequencing provides critical insights, yet a persistent gap remains in computational tools for detailed disease progression analysis and targeted in-silico drug interventions. Here, we introduce UNAGI, a deep generative neural network tailored to analyze time-series single-cell transcriptomic data. This tool captures the complex cellular dynamics underlying disease progression, enhancing drug perturbation modeling and discovery. When applied to a dataset from patients with Idiopathic Pulmonary Fibrosis (IPF), UNAGI learns disease-informed cell embeddings that sharpen our understanding of disease progression, leading to the identification of potential therapeutic drug candidates. Validation via proteomics reveals the accuracy of UNAGI's cellular dynamics analyses, and the use of the Fibrotic Cocktail treated human Precision-cut Lung Slices confirms UNAGI's predictions that Nifedipine, an antihypertensive drug, may have antifibrotic effects on human tissues. UNAGI's versatility extends to other diseases, including a COVID dataset, demonstrating adaptability and confirming its broader applicability in decoding complex cellular dynamics beyond IPF, amplifying its utility in the quest for therapeutic solutions across diverse pathological landscapes.
Collapse
Affiliation(s)
- Yumin Zheng
- Quantitative Life Sciences, Faculty of Medicine & Health Sciences, McGill University, Montreal, QC, Canada
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jonas C. Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Taylor Adams
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Aurelien Justet
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Farida Ahangari
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Xiting Yan
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Paul Hansen
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Marianne Carlon
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Emanuela Cortesi
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Laurens J. De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Ivan O Rosas
- Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ricardo Pineda
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John Sembrat
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melanie Königshoff
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John E. McDonough
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Wim A. Wuyts
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, United States
| | - Jun Ding
- Quantitative Life Sciences, Faculty of Medicine & Health Sciences, McGill University, Montreal, QC, Canada
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Mila - Quebec AI Institute, Montreal, QC, Canada
| |
Collapse
|
3
|
Ren JX, Gao Q, Zhou XC, Chen L, Guo W, Feng KY, Lu L, Huang T, Cai YD. Identification of Gene Markers Associated with COVID-19 Severity and Recovery in Different Immune Cell Subtypes. BIOLOGY 2023; 12:947. [PMID: 37508378 PMCID: PMC10376631 DOI: 10.3390/biology12070947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023]
Abstract
As COVID-19 develops, dynamic changes occur in the patient's immune system. Changes in molecular levels in different immune cells can reflect the course of COVID-19. This study aims to uncover the molecular characteristics of different immune cell subpopulations at different stages of COVID-19. We designed a machine learning workflow to analyze scRNA-seq data of three immune cell types (B, T, and myeloid cells) in four levels of COVID-19 severity/outcome. The datasets for three cell types included 403,700 B-cell, 634,595 T-cell, and 346,547 myeloid cell samples. Each cell subtype was divided into four groups, control, convalescence, progression mild/moderate, and progression severe/critical, and each immune cell contained 27,943 gene features. A feature analysis procedure was applied to the data of each cell type. Irrelevant features were first excluded according to their relevance to the target variable measured by mutual information. Then, four ranking algorithms (last absolute shrinkage and selection operator, light gradient boosting machine, Monte Carlo feature selection, and max-relevance and min-redundancy) were adopted to analyze the remaining features, resulting in four feature lists. These lists were fed into the incremental feature selection, incorporating three classification algorithms (decision tree, k-nearest neighbor, and random forest) to extract key gene features and construct classifiers with superior performance. The results confirmed that genes such as PFN1, RPS26, and FTH1 played important roles in SARS-CoV-2 infection. These findings provide a useful reference for the understanding of the ongoing effect of COVID-19 development on the immune system.
Collapse
Affiliation(s)
- Jing-Xin Ren
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Qian Gao
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiao-Chao Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China
| | - Kai-Yan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
4
|
Avila-Ponce de León U, Vázquez-Jiménez A, Padilla-Longoria P, Resendis-Antonio O. Uncoding the interdependency of tumor microenvironment and macrophage polarization: insights from a continuous network approach. Front Immunol 2023; 14:1150890. [PMID: 37283734 PMCID: PMC10240616 DOI: 10.3389/fimmu.2023.1150890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
The balance between pro- and anti-inflammatory immune system responses is crucial to preventing complex diseases like cancer. Macrophages are essential immune cells that contribute to this balance constrained by the local signaling profile of the tumor microenvironment. To understand how pro- and anti-inflammatory unbalance emerges in cancer, we developed a theoretical analysis of macrophage differentiation that is derived from activated monocytes circulating in the blood. Once recruited to the site of inflammation, monocytes can be polarized based on the specific interleukins and chemokines in the microenvironment. To quantify this process, we used a previous regulatory network reconstructed by our group and transformed Boolean Network attractors of macrophage polarization to an ODE scheme, it enables us to quantify the activation of their genes in a continuous fashion. The transformation was developed using the interaction rules with a fuzzy logic approach. By implementing this approach, we analyzed different aspects that cannot be visualized in the Boolean setting. For example, this approach allows us to explore the dynamic behavior at different concentrations of cytokines and transcription factors in the microenvironment. One important aspect to assess is the evaluation of the transitions between phenotypes, some of them characterized by an abrupt or a gradual transition depending on specific concentrations of exogenous cytokines in the tumor microenvironment. For instance, IL-10 can induce a hybrid state that transits between an M2c and an M2b macrophage. Interferon- γ can induce a hybrid between M1 and M1a macrophage. We further demonstrated the plasticity of macrophages based on a combination of cytokines and the existence of hybrid phenotypes or partial polarization. This mathematical model allows us to unravel the patterns of macrophage differentiation based on the competition of expression of transcriptional factors. Finally, we survey how macrophages may respond to a continuously changing immunological response in a tumor microenvironment.
Collapse
Affiliation(s)
- Ugo Avila-Ponce de León
- Programa de Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de Mexico, Mexico
| | - Aarón Vázquez-Jiménez
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de Mexico, Mexico
| | - Pablo Padilla-Longoria
- Institute for Applied Mathematics (IIMAS), Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de Mexico, Mexico
- Coordinación de la Investigación Científica - Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico, Mexico
- Centro de Ciencias de la Complejidad (C3), Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| |
Collapse
|
5
|
Romo-Rodríguez R, Gutiérrez-de Anda K, López-Blanco JA, Zamora-Herrera G, Cortés-Hernández P, Santos-López G, Márquez-Domínguez L, Vilchis-Ordoñez A, Ramírez-Ramírez D, Balandrán JC, Parra-Ortega I, Resendis-Antonio O, Domínguez-Ramírez L, López-Macías C, Bonifaz LC, Arriaga-Pizano LA, Cérbulo-Vázquez A, Ferat-Osorio E, Chavez-González A, Treviño S, Brambila E, Ramos-Sánchez MÁ, Toledo-Tapia R, Domínguez F, Bayrán-Flores J, Cruz-Oseguera A, Reyes-Leyva JR, Méndez-Martínez S, Ayón-Aguilar J, Treviño-García A, Monjaraz E, Pelayo R. Chronic Comorbidities in Middle Aged Patients Contribute to Ineffective Emergency Hematopoiesis in Covid-19 Fatal Outcomes. Arch Med Res 2023; 54:197-210. [PMID: 36990888 PMCID: PMC10015105 DOI: 10.1016/j.arcmed.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/21/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Background and Aims . Mexico is among the countries with the highest estimated excess mortality rates due to the COVID–19 pandemic, with more than half of reported deaths occurring in adults younger than 65 years old. Although this behavior is presumably influenced by the young demographics and the high prevalence of metabolic diseases, the underlying mechanisms have not been determined. Methods . The age–stratified case fatality rate (CFR) was estimated in a prospective cohort with 245 hospitalized COVID–19 cases, followed through time, for the period October 2020–September 2021. Cellular and inflammatory parameters were exhaustively investigated in blood samples by laboratory test, multiparametric flow cytometry and multiplex immunoassays. Results . The CFR was 35.51%, with 55.2% of deaths recorded in middle–aged adults. On admission, hematological cell differentiation, physiological stress and inflammation parameters, showed distinctive profiles of potential prognostic value in patients under 65 at 7 d follow–up. Pre–existing metabolic conditions were identified as risk factors of poor outcomes. Chronic kidney disease (CKD), as single comorbidity or in combination with diabetes, had the highest risk for COVID–19 fatality. Of note, fatal outcomes in middle–aged patients were marked from admission by an inflammatory landscape and emergency myeloid hematopoiesis at the expense of functional lymphoid innate cells for antiviral immunosurveillance, including NK and dendritic cell subsets. Conclusions . Comorbidities increased the development of imbalanced myeloid phenotype, rendering middle–aged individuals unable to effectively control SARS–CoV–2. A predictive signature of high–risk outcomes at day 7 of disease evolution as a tool for their early stratification in vulnerable populations is proposed.
Collapse
Affiliation(s)
- Rubí Romo-Rodríguez
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México; Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Karla Gutiérrez-de Anda
- Hospital General de Zona 5, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Jebea A López-Blanco
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Gabriela Zamora-Herrera
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México; Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Paulina Cortés-Hernández
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Gerardo Santos-López
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Luis Márquez-Domínguez
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | | | - Dalia Ramírez-Ramírez
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | | | - Israel Parra-Ortega
- Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México, México
| | - Osbaldo Resendis-Antonio
- Instituto Nacional de Medicina Genómica (INMEGEN) & Coordinación de la Investigación Científica-Red de Apoyo a la Investigación-Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | | | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Laura C Bonifaz
- Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Lourdes A Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | | | - Eduardo Ferat-Osorio
- Dirección de Educación e Investigación en Salud, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Antonieta Chavez-González
- Unidad de Investigación Médica en Enfermedades Oncológicas, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla. Puebla, Mexico
| | - Eduardo Brambila
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla. Puebla, Mexico
| | - Miguel Ángel Ramos-Sánchez
- Unidad de Medicina Familiar 57. Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México; Hospital General de Zona 20, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Ricardo Toledo-Tapia
- Hospital General de Zona 20, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Fabiola Domínguez
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Jorge Bayrán-Flores
- Hospital General de Zona 5, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Alejandro Cruz-Oseguera
- Hospital General de Zona 5, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Julio Roberto Reyes-Leyva
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México
| | - Socorro Méndez-Martínez
- Coordinación de Planeación y Enlace Institucional, Instituto Mexicano del Seguro Social, Delegación Puebla, México
| | - Jorge Ayón-Aguilar
- Coordinación Médica de Investigación en Salud, Instituto Mexicano del Seguro Social, Delegación Puebla, México
| | - Aurora Treviño-García
- Órganos de Operación Administrativa Desconcentrada, Instituto Mexicano del Seguro Social, Delegación Puebla, México
| | - Eduardo Monjaraz
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Rosana Pelayo
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Puebla, México; Unidad de Educación e Investigación, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| |
Collapse
|
6
|
Avila-Ponce de León U, Vazquez-Jimenez A, Cervera A, Resendis-González G, Neri-Rosario D, Resendis-Antonio O. Machine Learning and COVID-19: Lessons from SARS-CoV-2. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:311-335. [PMID: 37378775 DOI: 10.1007/978-3-031-28012-2_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Currently, methods in machine learning have opened a significant number of applications to construct classifiers with capacities to recognize, identify, and interpret patterns hidden in massive amounts of data. This technology has been used to solve a variety of social and health issues against coronavirus disease 2019 (COVID-19). In this chapter, we present some supervised and unsupervised machine learning techniques that have contributed in three aspects to supplying information to health authorities and diminishing the deadly effects of the current worldwide outbreak on the population. First is the identification and construction of powerful classifiers capable of predicting severe, moderate, or asymptomatic responses in COVID-19 patients starting from clinical or high-throughput technologies. Second is the identification of groups of patients with similar physiological responses to improve the triage classification and inform treatments. The final aspect is the combination of machine learning methods and schemes from systems biology to link associative studies with mechanistic frameworks. This chapter aims to discuss some practical applications in the use of machine learning techniques to handle data coming from social behavior and high-throughput technologies, associated with COVID-19 evolution.
Collapse
Affiliation(s)
- Ugo Avila-Ponce de León
- Programa de Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México, Mexico
| | - Aarón Vazquez-Jimenez
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México, Mexico
| | - Alejandra Cervera
- Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México, Mexico
| | - Galilea Resendis-González
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México, Mexico
| | - Daniel Neri-Rosario
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México, Mexico
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México, Mexico.
- Coordinación de la Investigación Científica - Red de Apoyo a la Investigación - Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico.
| |
Collapse
|
7
|
Ratnasiri K, Wilk AJ, Lee MJ, Khatri P, Blish CA. Single-cell RNA-seq methods to interrogate virus-host interactions. Semin Immunopathol 2023; 45:71-89. [PMID: 36414692 PMCID: PMC9684776 DOI: 10.1007/s00281-022-00972-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022]
Abstract
The twenty-first century has seen the emergence of many epidemic and pandemic viruses, with the most recent being the SARS-CoV-2-driven COVID-19 pandemic. As obligate intracellular parasites, viruses rely on host cells to replicate and produce progeny, resulting in complex virus and host dynamics during an infection. Single-cell RNA sequencing (scRNA-seq), by enabling broad and simultaneous profiling of both host and virus transcripts, represents a powerful technology to unravel the delicate balance between host and virus. In this review, we summarize technological and methodological advances in scRNA-seq and their applications to antiviral immunity. We highlight key scRNA-seq applications that have enabled the understanding of viral genomic and host response heterogeneity, differential responses of infected versus bystander cells, and intercellular communication networks. We expect further development of scRNA-seq technologies and analytical methods, combined with measurements of additional multi-omic modalities and increased availability of publicly accessible scRNA-seq datasets, to enable a better understanding of viral pathogenesis and enhance the development of antiviral therapeutics strategies.
Collapse
Affiliation(s)
- Kalani Ratnasiri
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Aaron J Wilk
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Madeline J Lee
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Purvesh Khatri
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Medicine, Center for Biomedical Informatics Research, Stanford, CA, USA.
- Inflammatix, Inc., Sunnyvale, CA, 94085, USA.
| | - Catherine A Blish
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
8
|
de León UAP, Resendis-Antonio O. Macrophage Boolean networks in the time of SARS-CoV-2. Front Immunol 2022; 13:997434. [DOI: 10.3389/fimmu.2022.997434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
|
9
|
Samy A, Maher MA, Abdelsalam NA, Badr E. SARS-CoV-2 potential drugs, drug targets, and biomarkers: a viral-host interaction network-based analysis. Sci Rep 2022; 12:11934. [PMID: 35831333 PMCID: PMC9279364 DOI: 10.1038/s41598-022-15898-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/30/2022] [Indexed: 12/13/2022] Open
Abstract
COVID-19 is a global pandemic impacting the daily living of millions. As variants of the virus evolve, a complete comprehension of the disease and drug targets becomes a decisive duty. The Omicron variant, for example, has a notably high transmission rate verified in 155 countries. We performed integrative transcriptomic and network analyses to identify drug targets and diagnostic biomarkers and repurpose FDA-approved drugs for SARS-CoV-2. Upon the enrichment of 464 differentially expressed genes, pathways regulating the host cell cycle were significant. Regulatory and interaction networks featured hsa-mir-93-5p and hsa-mir-17-5p as blood biomarkers while hsa-mir-15b-5p as an antiviral agent. MYB, RRM2, ERG, CENPF, CIT, and TOP2A are potential drug targets for treatment. HMOX1 is suggested as a prognostic biomarker. Enhancing HMOX1 expression by neem plant extract might be a therapeutic alternative. We constructed a drug-gene network for FDA-approved drugs to be repurposed against the infection. The key drugs retrieved were members of anthracyclines, mitotic inhibitors, anti-tumor antibiotics, and CDK1 inhibitors. Additionally, hydroxyquinone and digitoxin are potent TOP2A inhibitors. Hydroxyurea, cytarabine, gemcitabine, sotalol, and amiodarone can also be redirected against COVID-19. The analysis enforced the repositioning of fluorouracil and doxorubicin, especially that they have multiple drug targets, hence less probability of resistance.
Collapse
Affiliation(s)
- Asmaa Samy
- University of Science and Technology, Zewail City, Giza, 12578, Egypt
| | - Mohamed A Maher
- University of Science and Technology, Zewail City, Giza, 12578, Egypt
| | - Nehal Adel Abdelsalam
- University of Science and Technology, Zewail City, Giza, 12578, Egypt.,Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Eman Badr
- University of Science and Technology, Zewail City, Giza, 12578, Egypt. .,Faculty of Computers and Artificial Intelligence, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
10
|
López-Cortés GI, Palacios-Pérez M, Veledíaz HF, Hernández-Aguilar M, López-Hernández GR, Zamudio GS, José MV. The Spike Protein of SARS-CoV-2 Is Adapting Because of Selective Pressures. Vaccines (Basel) 2022; 10:864. [PMID: 35746472 PMCID: PMC9230601 DOI: 10.3390/vaccines10060864] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/14/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
The global scale of the COVID-19 pandemic has demonstrated the evolution of SARS-CoV-2 and the clues of adaptation. After two years and two months since the declaration of the pandemic, several variants have emerged and become fixed in the human population thanks to extrinsic selective pressures but also to the inherent mutational capacity of the virus. Here, we applied a neutral substitution evolution test to the spike (S) protein of Omicron's protein and compared it to the others' variant of concern (VOC) neutral evolution. We carried out comparisons among the interactions between the S proteins from the VOCs (Alpha, Beta, Gamma, Delta and Omicron) and the receptor ACE2. The shared amino acids among all the ACE2 binding S proteins remain constant, indicating that these amino acids are essential for the accurate binding to the receptor. The complexes of the RBD for every variant with the receptor were used to identify the amino acids involved in the protein-protein interaction (PPI). The RBD of Omicron establishes 82 contacts, compared to the 74 of the Wuhan original viral protein. Hence, the mean number of contacts per residue is higher, making the contact thermodynamically more stable. The RBDs of the VOCs are similar in sequence and structure; however, Omicron's RBD presents the largest deviation from the structure by 1.11 Å RMSD, caused by a set of mutations near the glycosylation N343. The chemical properties and structure near the glycosylation N343 of the Omicron S protein are different from the original protein, which provoke reduced recognition by the neutralizing antibodies. Our results hint that selective pressures are induced by mass vaccination throughout the world and by the persistence of recurrent infections in immunosuppressed individuals, who did not eliminate the infection and ended up facilitating the selection of viruses whose characteristics are different from the previous VOCs, less pathogenic but with higher transmissibility.
Collapse
Affiliation(s)
- Georgina I. López-Cortés
- Theoretical Biology Group, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.P.-P.); (H.F.V.); (M.H.-A.); (G.R.L.-H.); (G.S.Z.)
- Network of Researchers on the Chemical Evolution of Life, NoRCEL, Leeds LS7 3RB, UK
| | - Miryam Palacios-Pérez
- Theoretical Biology Group, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.P.-P.); (H.F.V.); (M.H.-A.); (G.R.L.-H.); (G.S.Z.)
- Network of Researchers on the Chemical Evolution of Life, NoRCEL, Leeds LS7 3RB, UK
| | - Hannya F. Veledíaz
- Theoretical Biology Group, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.P.-P.); (H.F.V.); (M.H.-A.); (G.R.L.-H.); (G.S.Z.)
| | - Margarita Hernández-Aguilar
- Theoretical Biology Group, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.P.-P.); (H.F.V.); (M.H.-A.); (G.R.L.-H.); (G.S.Z.)
| | - Gerardo R. López-Hernández
- Theoretical Biology Group, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.P.-P.); (H.F.V.); (M.H.-A.); (G.R.L.-H.); (G.S.Z.)
| | - Gabriel S. Zamudio
- Theoretical Biology Group, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.P.-P.); (H.F.V.); (M.H.-A.); (G.R.L.-H.); (G.S.Z.)
| | - Marco V. José
- Theoretical Biology Group, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.P.-P.); (H.F.V.); (M.H.-A.); (G.R.L.-H.); (G.S.Z.)
- Network of Researchers on the Chemical Evolution of Life, NoRCEL, Leeds LS7 3RB, UK
| |
Collapse
|
11
|
Dagur P, Rakshit G, Sheikh M, Biswas A, Jha P, Al-Khafaji K, Ghosh M. Target prediction, computational identification, and network-based pharmacology of most potential phytoconstituent in medicinal leaves of Justicia adhatoda against SARS-CoV-2. J Biomol Struct Dyn 2022; 41:3926-3942. [PMID: 35412437 DOI: 10.1080/07391102.2022.2059010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The current global epidemic of the novel coronavirus (SARS-CoV-2) has been labeled a global public health emergency since it is causing substantial morbidity and mortality on daily basis. We need to identify an effective medication against SARS-CoV-2 because of its fast dissemination and re-emergence. This research is being carried out as part of a larger strategy to identify the most promising therapeutic targets using protein-protein interactions analysis. Mpro has been identified as one of the most important therapeutic targets. In this study, we did in-silico investigations to identify the target and further molecular docking, ADME, and toxicity prediction were done to assess the potential phyto-active antiviral compounds from Justicia adhatoda as powerful inhibitors of the Mpro of SARS-COV-2. We also investigated the capacity of these molecules to create stable interactions with the Mpro using 100 ns molecular dynamics simulation. The highest scoring compounds (taraxerol, friedelanol, anisotine, and adhatodine) were also found to exhibit excellent solubility and pharmacodynamic characteristics. We employed MMPBSA simulations to assess the stability of docked molecules in the Mpro binding site, revealing that the above compounds form the most stable complex with the Mpro. Network-based Pharmacology suggested that the selected compounds have various modes of action against SARS-CoV-2 that include immunoreaction enrichment, inflammatory reaction suppression, and more. These findings point to a promising class of drugs that should be investigated further in biochemical and cell-based studies to see their effectiveness against nCOVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pankaj Dagur
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, Ranchi, India
| | - Gourav Rakshit
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, Ranchi, India
| | - Murtuja Sheikh
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, Ranchi, India
| | - Abanish Biswas
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, Ranchi, India
| | - Parineeta Jha
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, Ranchi, India
| | - Khattab Al-Khafaji
- Department of Medical Laboratory Technology, Al-Nisour University College, Baghdad, Iraq
| | - Manik Ghosh
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, Ranchi, India
| |
Collapse
|