1
|
Gremese E, Tolusso B, Bruno D, Paglionico AM, Perniola S, Ferraccioli G, Alivernini S. COVID-19 illness: Different comorbidities may require different immunological therapeutic targets. Eur J Clin Invest 2023; 53:e14096. [PMID: 37724937 DOI: 10.1111/eci.14096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/26/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND The SARS-CoV-2 pandemic has led to more than 6,870.000 deaths worldwide. Despite recent therapeutic advances, deaths in Intensive Care Units still range between 34 and 72%, comprising substantial unmet need as we move to an endemic phase. The general agreement is that in the first few days of infection, antiviral drugs and neutralizing monoclonal antibodies should be adopted. When the patient is hospitalized and develops severe pneumonia, progressing to a systemic disease, immune modifying therapy with corticosteroids is indicated. Such interventions, however, are less effective in the context of comorbidities (e.g., diabetes, hypertension, heart failure, atrial fibrillation, obesity and central nervous system-CNS diseases) which are by themselves associated with poor outcomes. Such comorbidities comprise common and some distinct underlying inflammatory pathobiology regulated by differential cytokine taxonomy. METHODS Searching in the PubMed database, literature pertaining to the biology underlying the different comorbidities, and the data from the studies related to various immunological treatments for the Covid-19 disease were carefully analyzed. RESULTS Several experimental and clinical data have demonstrated that hypertension and atrial fibrillation present an IL-6 dependent signature, whereas diabetes, obesity, heart failure and CNS diseases may exhibit an IL-1a/b predominant signature. Distinct selective cytokine targeting may offer advantage in treating severe COVID-19 illness based on single or multiple associated comorbidities. When the patient does not immediately respond, a broader target range through JAKs pathway inhibitors may be indicated. CONCLUSIONS Herein, we discuss the biological background associated with distinct comorbidities which might impact the SARS-CoV-2 infection course and how these should to be addressed to improve the current therapeutic outcome.
Collapse
Affiliation(s)
- Elisa Gremese
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
- Immunology Core Facility, GSTEP, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Barbara Tolusso
- Immunology Core Facility, GSTEP, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Dario Bruno
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Anna Maria Paglionico
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Simone Perniola
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | | | - Stefano Alivernini
- Catholic University of the Sacred Heart, Rome, Italy
- Immunology Core Facility, GSTEP, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Rheumatology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
2
|
Taheri G, Habibi M. Identification of essential genes associated with SARS-CoV-2 infection as potential drug target candidates with machine learning algorithms. Sci Rep 2023; 13:15141. [PMID: 37704748 PMCID: PMC10499814 DOI: 10.1038/s41598-023-42127-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires the fast discovery of effective treatments to fight this worldwide concern. Several genes associated with the SARS-CoV-2, which are essential for its functionality, pathogenesis, and survival, have been identified. These genes, which play crucial roles in SARS-CoV-2 infection, are considered potential therapeutic targets. Developing drugs against these essential genes to inhibit their regular functions could be a good approach for COVID-19 treatment. Artificial intelligence and machine learning methods provide powerful infrastructures for interpreting and understanding the available data and can assist in finding fast explanations and cures. We propose a method to highlight the essential genes that play crucial roles in SARS-CoV-2 pathogenesis. For this purpose, we define eleven informative topological and biological features for the biological and PPI networks constructed on gene sets that correspond to COVID-19. Then, we use three different unsupervised learning algorithms with different approaches to rank the important genes with respect to our defined informative features. Finally, we present a set of 18 important genes related to COVID-19. Materials and implementations are available at: https://github.com/MahnazHabibi/Gene_analysis .
Collapse
Affiliation(s)
- Golnaz Taheri
- Department of Computer and Systems Sciences, Stockholm University, Stockholm, Sweden.
- Science for Life Laboratory, Stockholm, Sweden.
| | - Mahnaz Habibi
- Department of Mathematics, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| |
Collapse
|
3
|
Grassi M, Tarantino B. SEMtree: tree-based structure learning methods with structural equation models. Bioinformatics 2023; 39:btad377. [PMID: 37294820 PMCID: PMC10287946 DOI: 10.1093/bioinformatics/btad377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 05/08/2023] [Accepted: 06/08/2023] [Indexed: 06/11/2023] Open
Abstract
MOTIVATION With the exponential growth of expression and protein-protein interaction (PPI) data, the identification of functional modules in PPI networks that show striking changes in molecular activity or phenotypic signatures becomes of particular interest to reveal process-specific information that is correlated with cellular or disease states. This requires both the identification of network nodes with reliability scores and the availability of an efficient technique to locate the network regions with the highest scores. In the literature, a number of heuristic methods have been suggested. We propose SEMtree(), a set of tree-based structure discovery algorithms, combining graph and statistically interpretable parameters together with a user-friendly R package based on structural equation models framework. RESULTS Condition-specific changes from differential expression and gene-gene co-expression are recovered with statistical testing of node, directed edge, and directed path difference between groups. In the end, from a list of seed (i.e. disease) genes or gene P-values, the perturbed modules with undirected edges are generated with five state-of-the-art active subnetwork detection methods. The latter are supplied to causal additive trees based on Chu-Liu-Edmonds' algorithm (Chow and Liu, Approximating discrete probability distributions with dependence trees. IEEE Trans Inform Theory 1968;14:462-7) in SEMtree() to be converted in directed trees. This conversion allows to compare the methods in terms of directed active subnetworks. We applied SEMtree() to both Coronavirus disease (COVID-19) RNA-seq dataset (GEO accession: GSE172114) and simulated datasets with various differential expression patterns. Compared to existing methods, SEMtree() is able to capture biologically relevant subnetworks with simple visualization of directed paths, good perturbation extraction, and classifier performance. AVAILABILITY AND IMPLEMENTATION SEMtree() function is implemented in the R package SEMgraph, easily available at https://CRAN.R-project.org/package=SEMgraph.
Collapse
Affiliation(s)
- Mario Grassi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - Barbara Tarantino
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| |
Collapse
|
4
|
Barh D, Aburjaile FF, Tavares TS, da Silva ME, Bretz GPM, Rocha IFM, Dey A, de Souza RP, Góes-Neto A, Ribeiro SP, Alzahrani KJ, Alghamdi AA, Alzahrani FM, Halawani IF, Tiwari S, Aljabali AAA, Lundstrom K, Azevedo V, Ganguly NK. Indian food habit & food ingredients may have a role in lowering the severity & high death rate from COVID-19 in Indians: findings from the first nutrigenomic analysis. Indian J Med Res 2023; 157:293-303. [PMID: 37102510 PMCID: PMC10438415 DOI: 10.4103/ijmr.ijmr_1701_22] [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: 08/01/2022] [Indexed: 04/28/2023] Open
Abstract
Background & objectives During the COVID-19 pandemic, the death rate was reportedly 5-8 fold lower in India which is densely populated as compared to less populated western countries. The aim of this study was to investigate whether dietary habits were associated with the variations in COVID-19 severity and deaths between western and Indian population at the nutrigenomics level. Methods In this study nutrigenomics approach was applied. Blood transcriptome of severe COVID-19 patients from three western countries (showing high fatality) and two datasets from Indian patients were used. Gene set enrichment analyses were performed for pathways, metabolites, nutrients, etc., and compared for western and Indian samples to identify the food- and nutrient-related factors, which may be associated with COVID-19 severity. Data on the daily consumption of twelve key food components across four countries were collected and a correlation between nutrigenomics analyses and per capita daily dietary intake was investigated. Results Distinct dietary habits of Indians were observed, which may be associated with low death rate from COVID-19. Increased consumption of red meat, dairy products and processed foods by western populations may increase the severity and death rate by activating cytokine storm-related pathways, intussusceptive angiogenesis, hypercapnia and enhancing blood glucose levels due to high contents of sphingolipids, palmitic acid and byproducts such as CO2 and lipopolysaccharide (LPS). Palmitic acid also induces ACE2 expression and increases the infection rate. Coffee and alcohol that are highly consumed in western countries may increase the severity and death rates from COVID-19 by deregulating blood iron, zinc and triglyceride levels. The components of Indian diets maintain high iron and zinc concentrations in blood and rich fibre in their foods may prevent CO2 and LPS-mediated COVID-19 severity. Regular consumption of tea by Indians maintains high high-density lipoprotein (HDL) and low triglyceride in blood as catechins in tea act as natural atorvastatin. Importantly, regular consumption of turmeric in daily food by Indians maintains strong immunity and curcumin in turmeric may prevent pathways and mechanisms associated with SARS-CoV-2 infection and COVID-19 severity and lowered the death rate. Interpretation & conclusions Our results suggest that Indian food components suppress cytokine storm and various other severity related pathways of COVID-19 and may have a role in lowering severity and death rates from COVID-19 in India as compared to western populations. However, large multi-centered case-control studies are required to support our current findings.
Collapse
Affiliation(s)
- Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics & Applied Biotechnology, Purba Medinipur, West Bengal, India
- Department of Genetics, Ecology & Evolution, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Flávia Figueira Aburjaile
- Department of Preventative Veterinary Medicine, School of Veterinary Medicine, Belo Horizonte, Brazil
| | - Thais Silva Tavares
- Department of Laboratory of Algorithms in Biology, Institute of Biological Sciences, Belo Horizonte, Brazil
| | | | | | - Igor Fernando Martins Rocha
- Department of Centre of Research on Health Vulnerability, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Annesha Dey
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics & Applied Biotechnology, Purba Medinipur, West Bengal, India
| | - Renan Pedra de Souza
- Department of Laboratory of Integrative Biology, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Aristóteles Góes-Neto
- Department of Genetics, Ecology & Evolution, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Sérvio Pontes Ribeiro
- Department of Laboratory of Ecology of Diseases & Forests, Nucleus of Biological Research, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Khalid J. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ahmad A. Alghamdi
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Fuad M. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ibrahim Faisal Halawani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Sandeep Tiwari
- Department of Post-Graduation Programs in Microbiology and Immunology, Institute of Biology and Health Sciences, Federal University of Bahia, Salvador, BA, Brazil
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics & Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | | | - Vasco Azevedo
- Department of Genetics, Ecology & Evolution, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Nirmal Kumar Ganguly
- Policy Center for Biomedical Research, Translational Health Science & Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
5
|
Lundstrom K, Hromić-Jahjefendić A, Bilajac E, Aljabali AAA, Baralić K, Sabri NA, Shehata EM, Raslan M, Ferreira ACBH, Orlandi L, Serrano-Aroca Á, Tambuwala MM, Uversky VN, Azevedo V, Alzahrani KJ, Alsharif KF, Halawani IF, Alzahrani FM, Redwan EM, Barh D. COVID-19 signalome: Pathways for SARS-CoV-2 infection and impact on COVID-19 associated comorbidity. Cell Signal 2023; 101:110495. [PMID: 36252792 PMCID: PMC9568271 DOI: 10.1016/j.cellsig.2022.110495] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has been the focus of research the past two years. The major breakthrough was made by discovering pathways related to SARS-CoV-2 infection through cellular interaction by angiotensin-converting enzyme (ACE2) and cytokine storm. The presence of ACE2 in lungs, intestines, cardiovascular tissues, brain, kidneys, liver, and eyes shows that SARS-CoV-2 may have targeted these organs to further activate intracellular signalling pathways that lead to cytokine release syndrome. It has also been reported that SARS-CoV-2 can hijack coatomer protein-I (COPI) for S protein retrograde trafficking to the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), which, in turn, acts as the assembly site for viral progeny. In infected cells, the newly synthesized S protein in endoplasmic reticulum (ER) is transported first to the Golgi body, and then from the Golgi body to the ERGIC compartment resulting in the formation of specific a motif at the C-terminal end. This review summarizes major events of SARS-CoV-2 infection route, immune response following host-cell infection as an important factor for disease outcome, as well as comorbidity issues of various tissues and organs arising due to COVID-19. Investigations on alterations of host-cell machinery and viral interactions with multiple intracellular signaling pathways could represent a major factor in more effective disease management.
Collapse
Affiliation(s)
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Esma Bilajac
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan.
| | - Katarina Baralić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Nagwa A Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11865, Egypt.
| | - Eslam M Shehata
- Drug Research Center, Clinical Research and Bioanalysis Department, Cairo 11865, Egypt.
| | - Mohamed Raslan
- Drug Research Center, Clinical Research and Bioanalysis Department, Cairo 11865, Egypt.
| | - Ana Cláudia B H Ferreira
- Campinas State University, Campinas, São Paulo, Brazil; University Center of Lavras (UNILAVRAS), Lavras, Minas Gerais, Brazil.
| | - Lidiane Orlandi
- University Center of Lavras (UNILAVRAS), Lavras, Minas Gerais, Brazil.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain.
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Vasco Azevedo
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Khalaf F Alsharif
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Ibrahim F Halawani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Fuad M Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia.
| | - Debmalya Barh
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, India.
| |
Collapse
|
6
|
Wang Z, Zhan J, Gao H. Computer-aided drug design combined network pharmacology to explore anti-SARS-CoV-2 or anti-inflammatory targets and mechanisms of Qingfei Paidu Decoction for COVID-19. Front Immunol 2022; 13:1015271. [PMID: 36618410 PMCID: PMC9816407 DOI: 10.3389/fimmu.2022.1015271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Coronavirus Disease-2019 (COVID-19) is an infectious disease caused by SARS-CoV-2. Severe cases of COVID-19 are characterized by an intense inflammatory process that may ultimately lead to organ failure and patient death. Qingfei Paidu Decoction (QFPD), a traditional Chines e medicine (TCM) formula, is widely used in China as anti-SARS-CoV-2 and anti-inflammatory. However, the potential targets and mechanisms for QFPD to exert anti-SARS-CoV-2 or anti-inflammatory effects remain unclear. Methods In this study, Computer-Aided Drug Design was performed to identify the antiviral or anti-inflammatory components in QFPD and their targets using Discovery Studio 2020 software. We then investigated the mechanisms associated with QFPD for treating COVID-19 with the help of multiple network pharmacology approaches. Results and discussion By overlapping the targets of QFPD and COVID-19, we discovered 8 common targets (RBP4, IL1RN, TTR, FYN, SFTPD, TP53, SRPK1, and AKT1) of 62 active components in QFPD. These may represent potential targets for QFPD to exert anti-SARS-CoV-2 or anti-inflammatory effects. The result showed that QFPD might have therapeutic effects on COVID-19 by regulating viral infection, immune and inflammation-related pathways. Our work will promote the development of new drugs for COVID-19.
Collapse
Affiliation(s)
| | | | - Hongwei Gao
- School of Life Science, Ludong University, Yantai, Shandong, China
| |
Collapse
|
7
|
Bu F, Guan R, Wang W, Liu Z, Yin S, Zhao Y, Chai J. Bioinformatics and systems biology approaches to identify the effects of COVID-19 on neurodegenerative diseases: A review. Medicine (Baltimore) 2022; 101:e32100. [PMID: 36626425 PMCID: PMC9750669 DOI: 10.1097/md.0000000000032100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing coronavirus disease (COVID-19), has been devastated by COVID-19 in an increasing number of countries and health care systems around the world since its announcement of a global pandemic on 11 March 2020. During the pandemic, emerging novel viral mutant variants have caused multiple outbreaks of COVID-19 around the world and are prone to genetic evolution, causing serious damage to human health. As confirmed cases of COVID-19 spread rapidly, there is evidence that SARS-CoV-2 infection involves the central nervous system (CNS) and peripheral nervous system (PNS), directly or indirectly damaging neurons and further leading to neurodegenerative diseases (ND), but the molecular mechanisms of ND and CVOID-19 are unknown. We employed transcriptomic profiling to detect several major diseases of ND: Alzheimer 's disease (AD), Parkinson' s disease (PD), and multiple sclerosis (MS) common pathways and molecular biomarkers in association with COVID-19, helping to understand the link between ND and COVID-19. There were 14, 30 and 19 differentially expressed genes (DEGs) between COVID-19 and Alzheimer 's disease (AD), Parkinson' s disease (PD) and multiple sclerosis (MS), respectively; enrichment analysis showed that MAPK, IL-17, PI3K-Akt and other signaling pathways were significantly expressed; the hub genes (HGs) of DEGs between ND and COVID-19 were CRH, SST, TAC1, SLC32A1, GAD2, GAD1, VIP and SYP. Analysis of transcriptome data suggests multiple co-morbid mechanisms between COVID-19 and AD, PD, and MS, providing new ideas and therapeutic strategies for clinical prevention and treatment of COVID-19 and ND.
Collapse
Affiliation(s)
- Fan Bu
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
- * Correspondence: Fan Bu, Heilongjiang University of Chinese Medicine, Haerbin 150040, Heilongjiang Province, China (e-mail: )
| | - Ruiqian Guan
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
- Heilongjiang University of Chinese Medicine Affiliated Second Hospital, Haerbin, Heilongjiang Province, China
| | - Wanyu Wang
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| | - Zhao Liu
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| | - Shijie Yin
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| | - Yonghou Zhao
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
- Heilongjiang University of Chinese Medicine Affiliated Second Hospital, Haerbin, Heilongjiang Province, China
| | - Jianbo Chai
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| |
Collapse
|
8
|
Taheri G, Habibi M. Comprehensive analysis of pathways in Coronavirus 2019 (COVID-19) using an unsupervised machine learning method. Appl Soft Comput 2022; 128:109510. [PMID: 35992221 PMCID: PMC9384336 DOI: 10.1016/j.asoc.2022.109510] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/07/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022]
Abstract
The World Health Organization (WHO) introduced “Coronavirus disease 19” or “COVID-19” as a novel coronavirus in March 2020. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires the fast discovery of effective treatments to fight this worldwide crisis. Artificial intelligence and bioinformatics analysis pipelines can assist with finding biomarkers, explanations, and cures. Artificial intelligence and machine learning methods provide powerful infrastructures for interpreting and understanding the available data. On the other hand, pathway enrichment analysis, as a dominant tool, could help researchers discover potential key targets present in biological pathways of host cells that are targeted by SARS-CoV-2. In this work, we propose a two-stage machine learning approach for pathway analysis. During the first stage, four informative gene sets that can represent important COVID-19 related pathways are selected. These “representative genes” are associated with the COVID-19 pathology. Then, two distinctive networks were constructed for COVID-19 related signaling and disease pathways. In the second stage, the pathways of each network are ranked with respect to some unsupervised scorning method based on our defined informative features. Finally, we present a comprehensive analysis of the top important pathways in both networks. Materials and implementations are available at: https://github.com/MahnazHabibi/Pathway.
Collapse
Affiliation(s)
- Golnaz Taheri
- Department of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden.,Science for Life Laboratory, Stockholm, Sweden
| | - Mahnaz Habibi
- Department of Mathematics, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| |
Collapse
|
9
|
Xia J, Chen S, Li Y, Li H, Gan M, Wu J, Prohaska CC, Bai Y, Gao L, Gu L, Zhang D. Immune Response Is Key to Genetic Mechanisms of SARS-CoV-2 Infection With Psychiatric Disorders Based on Differential Gene Expression Pattern Analysis. Front Immunol 2022; 13:798538. [PMID: 35185890 PMCID: PMC8854505 DOI: 10.3389/fimmu.2022.798538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/13/2022] [Indexed: 12/11/2022] Open
Abstract
Existing evidence demonstrates that coronavirus disease 2019 (COVID-19) leads to psychiatric illness, despite its main clinical manifestations affecting the respiratory system. People with mental disorders are more susceptible to COVID-19 than individuals without coexisting mental health disorders, with significantly higher rates of severe illness and mortality in this population. The incidence of new psychiatric diagnoses after infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also remarkably high. SARS-CoV-2 has been reported to use angiotensin-converting enzyme-2 (ACE2) as a receptor for infecting susceptible cells and is expressed in various tissues, including brain tissue. Thus, there is an urgent need to investigate the mechanism linking psychiatric disorders to COVID-19. Using a data set of peripheral blood cells from patients with COVID-19, we compared this to data sets of whole blood collected from patients with psychiatric disorders and used bioinformatics and systems biology approaches to identify genetic links. We found a large number of overlapping immune-related genes between patients infected with SARS-CoV-2 and differentially expressed genes of bipolar disorder (BD), schizophrenia (SZ), and late-onset major depressive disorder (LOD). Many pathways closely related to inflammatory responses, such as MAPK, PPAR, and TGF-β signaling pathways, were observed by enrichment analysis of common differentially expressed genes (DEGs). We also performed a comprehensive analysis of protein-protein interaction network and gene regulation networks. Chemical-protein interaction networks and drug prediction were used to screen potential pharmacologic therapies. We hope that by elucidating the relationship between the pathogenetic processes and genetic mechanisms of infection with SARS-CoV-2 with psychiatric disorders, it will lead to innovative strategies for future research and treatment of psychiatric disorders linked to COVID-19.
Collapse
Affiliation(s)
- Jing Xia
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Shuhan Chen
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Yaping Li
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Hua Li
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Minghong Gan
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Jiashuo Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Clare Colette Prohaska
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Yang Bai
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lu Gao
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Li Gu
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Dongfang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
10
|
Hasankhani A, Bahrami A, Sheybani N, Aria B, Hemati B, Fatehi F, Ghaem Maghami Farahani H, Javanmard G, Rezaee M, Kastelic JP, Barkema HW. Differential Co-Expression Network Analysis Reveals Key Hub-High Traffic Genes as Potential Therapeutic Targets for COVID-19 Pandemic. Front Immunol 2021; 12:789317. [PMID: 34975885 PMCID: PMC8714803 DOI: 10.3389/fimmu.2021.789317] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background The recent emergence of COVID-19, rapid worldwide spread, and incomplete knowledge of molecular mechanisms underlying SARS-CoV-2 infection have limited development of therapeutic strategies. Our objective was to systematically investigate molecular regulatory mechanisms of COVID-19, using a combination of high throughput RNA-sequencing-based transcriptomics and systems biology approaches. Methods RNA-Seq data from peripheral blood mononuclear cells (PBMCs) of healthy persons, mild and severe 17 COVID-19 patients were analyzed to generate a gene expression matrix. Weighted gene co-expression network analysis (WGCNA) was used to identify co-expression modules in healthy samples as a reference set. For differential co-expression network analysis, module preservation and module-trait relationships approaches were used to identify key modules. Then, protein-protein interaction (PPI) networks, based on co-expressed hub genes, were constructed to identify hub genes/TFs with the highest information transfer (hub-high traffic genes) within candidate modules. Results Based on differential co-expression network analysis, connectivity patterns and network density, 72% (15 of 21) of modules identified in healthy samples were altered by SARS-CoV-2 infection. Therefore, SARS-CoV-2 caused systemic perturbations in host biological gene networks. In functional enrichment analysis, among 15 non-preserved modules and two significant highly-correlated modules (identified by MTRs), 9 modules were directly related to the host immune response and COVID-19 immunopathogenesis. Intriguingly, systemic investigation of SARS-CoV-2 infection identified signaling pathways and key genes/proteins associated with COVID-19's main hallmarks, e.g., cytokine storm, respiratory distress syndrome (ARDS), acute lung injury (ALI), lymphopenia, coagulation disorders, thrombosis, and pregnancy complications, as well as comorbidities associated with COVID-19, e.g., asthma, diabetic complications, cardiovascular diseases (CVDs), liver disorders and acute kidney injury (AKI). Topological analysis with betweenness centrality (BC) identified 290 hub-high traffic genes, central in both co-expression and PPI networks. We also identified several transcriptional regulatory factors, including NFKB1, HIF1A, AHR, and TP53, with important immunoregulatory roles in SARS-CoV-2 infection. Moreover, several hub-high traffic genes, including IL6, IL1B, IL10, TNF, SOCS1, SOCS3, ICAM1, PTEN, RHOA, GDI2, SUMO1, CASP1, IRAK3, HSPA5, ADRB2, PRF1, GZMB, OASL, CCL5, HSP90AA1, HSPD1, IFNG, MAPK1, RAB5A, and TNFRSF1A had the highest rates of information transfer in 9 candidate modules and central roles in COVID-19 immunopathogenesis. Conclusion This study provides comprehensive information on molecular mechanisms of SARS-CoV-2-host interactions and identifies several hub-high traffic genes as promising therapeutic targets for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Nuclear Agriculture Research School, Nuclear Science and Technology Research Institute, Karaj, Iran
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Behzad Aria
- Department of Physical Education and Sports Science, School of Psychology and Educational Sciences, Yazd University, Yazd, Iran
| | - Behzad Hemati
- Biotechnology Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Farhang Fatehi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | | | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mahsa Rezaee
- Department of Medical Mycology, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
11
|
Aljabali AAA, Pal K, Serrano-Aroca A, Takayama K, Dua K, Tambuwala MM. Clinical utility of novel biosensing platform: Diagnosis of coronavirus SARS-CoV-2 at point of care. MATERIALS LETTERS 2021; 304:130612. [PMID: 34381287 PMCID: PMC8343387 DOI: 10.1016/j.matlet.2021.130612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 06/13/2023]
Abstract
Early detection is the first step in the fight against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, an efficient, rapid, selective, specific, and inexpensive SARS-CoV-2 diagnostic method is the need of the hour. The reverse transcription-polymerase chain reaction (RT-PCR) technology is massively utilized to detect infection with SARS-CoV-2. However, scientists continue to strive to create enhanced technology while continually developing nanomaterial-enabled biosensing methods that can provide new methodologies, potentially fulfilling the present demand for rapid and early identification of coronavirus disease 2019 (COVID-19) patients. Our review presents a summary of the recent diagnosis of SARS-CoV-2 of COVID-19 pandemic and nanomaterial-available biosensing methods. Although limited research on nanomaterials-based nanosensors has been published, allowing for biosensing approaches for diagnosing SARS-CoV-2, this study highlights nanomaterials that provide an enhanced biosensing strategy and potential processes that lead to COVID-19 diagnosis.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University-Faculty of Pharmacy, Irbid 566, Jordan
| | - Kaushik Pal
- Federal University of Rio de Janeiro, Cidade Universitária, Laboratório de Biopolímeros e Sensores/LaBioS Centro de Tecnologia - Cidade Universitária, Rio de Janeiro, RJ 21941-901, Brazil
| | - Angel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Translational Research Centre San Alberto Magno, Catholic University of Valencia San Vicente M'artir, c/Guillem de Castro 94, 46001 Valencia, Spain
| | - Kazuo Takayama
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
| |
Collapse
|
12
|
Cell-Type-Specific Profibrotic Scores across Multi-Organ Systems Predict Cancer Prognosis. Cancers (Basel) 2021; 13:cancers13236024. [PMID: 34885134 PMCID: PMC8656778 DOI: 10.3390/cancers13236024] [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: 11/12/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Fibrosis is a major player and contributor in the tumor microenvironment. Profibrotic changes precede the early development and establishment of a variety of human diseases, such as fibrosis and cancer. Being able to measure such early signals at the single cell level is critically useful for identifying new mechanisms and potential drug targets for a wide range of diseases. This study was designed to computationally identify profibrotic cell populations using single-cell transcriptomic data and to identify gene signatures that could predict cancer prognosis. Abstract Fibrosis is a major cause of mortality. Key profibrotic mechanisms are common pathways involved in tumorigenesis. Characterizing the profibrotic phenotype will help reveal the underlying mechanisms of early development and progression of a variety of human diseases, such as fibrosis and cancer. Fibroblasts have been center stage in response to various stimuli, such as viral infections. However, a comprehensive catalog of cell types involved in this process is currently lacking. Here, we deployed single-cell transcriptomic data across multi-organ systems (i.e., heart, kidney, liver, and lung) to identify novel profibrotic cell populations based on ECM pathway activity at single-cell resolution. In addition to fibroblasts, we also reported that epithelial, endothelial, myeloid, natural killer T, and secretory cells, as well as proximal convoluted tubule cells of the nephron, were significantly actively involved. Cell-type-specific gene signatures were enriched in viral infection pathways, enhanced glycolysis, and carcinogenesis, among others; they were validated using independent datasets in this study. By projecting the signatures into bulk TCGA tumor samples, we could predict prognosis in the patients using profibrotic scores. Our profibrotic cellular phenotype is useful for identifying new mechanisms and potential drug targets at the cell-type level for a wide range of diseases involved in ECM pathway activation.
Collapse
|
13
|
Synowiec A, Jedrysik M, Branicki W, Klajmon A, Lei J, Owczarek K, Suo C, Szczepanski A, Wang J, Zhang P, Labaj PP, Pyrc K. Identification of Cellular Factors Required for SARS-CoV-2 Replication. Cells 2021; 10:cells10113159. [PMID: 34831382 PMCID: PMC8622730 DOI: 10.3390/cells10113159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/27/2021] [Accepted: 11/10/2021] [Indexed: 12/25/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the recently emerged virus responsible for the COVID-19 pandemic. Clinical presentation can range from asymptomatic disease and mild respiratory tract infection to severe disease with lung injury, multiorgan failure, and death. SARS-CoV-2 is the third animal coronavirus to emerge in humans in the 21st century, and coronaviruses appear to possess a unique ability to cross borders between species and infect a wide range of organisms. This is somewhat surprising as, except for the requirement of host cell receptors, cell–pathogen interactions are usually species-specific. Insights into these host–virus interactions will provide a deeper understanding of the process of SARS-CoV-2 infection and provide a means for the design and development of antiviral agents. In this study, we describe a complex analysis of SARS-CoV-2 infection using a genome-wide CRISPR-Cas9 knock-out system in HeLa cells overexpressing entry receptor angiotensin-converting enzyme 2 (ACE2). This platform allows for the identification of factors required for viral replication. This study was designed to include a high number of replicates (48 replicates; 16 biological repeats with 3 technical replicates each) to prevent data instability, remove sources of bias, and allow multifactorial bioinformatic analyses in order to study the resulting interaction network. The results obtained provide an interesting insight into the replication mechanisms of SARS-CoV-2.
Collapse
Affiliation(s)
- Aleksandra Synowiec
- ViroGenetics—BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (A.S.); (M.J.); (K.O.); (A.S.)
| | - Malwina Jedrysik
- ViroGenetics—BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (A.S.); (M.J.); (K.O.); (A.S.)
| | - Wojciech Branicki
- Human Genome Variation Research Group, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (W.B.); (A.K.)
| | - Adrianna Klajmon
- Human Genome Variation Research Group, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (W.B.); (A.K.)
| | - Jing Lei
- Key Laboratory of Public Health Safety, Department of Epidemiology & Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China; (J.L.); (C.S.); (J.W.); (P.Z.)
| | - Katarzyna Owczarek
- ViroGenetics—BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (A.S.); (M.J.); (K.O.); (A.S.)
| | - Chen Suo
- Key Laboratory of Public Health Safety, Department of Epidemiology & Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China; (J.L.); (C.S.); (J.W.); (P.Z.)
- Taizhou Institute of Health Sciences, Fudan University, Taizhou 225316, China
| | - Artur Szczepanski
- ViroGenetics—BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (A.S.); (M.J.); (K.O.); (A.S.)
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Jingru Wang
- Key Laboratory of Public Health Safety, Department of Epidemiology & Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China; (J.L.); (C.S.); (J.W.); (P.Z.)
| | - Pengyan Zhang
- Key Laboratory of Public Health Safety, Department of Epidemiology & Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China; (J.L.); (C.S.); (J.W.); (P.Z.)
| | - Pawel P. Labaj
- Bioinformatics Research Group, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland
- Correspondence: (P.P.L.); (K.P.)
| | - Krzysztof Pyrc
- ViroGenetics—BSL3 Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (A.S.); (M.J.); (K.O.); (A.S.)
- Correspondence: (P.P.L.); (K.P.)
| |
Collapse
|
14
|
Cano-Vicent A, Tuñón-Molina A, Martí M, Muramoto Y, Noda T, Takayama K, Serrano-Aroca Á. Antiviral Face Mask Functionalized with Solidified Hand Soap: Low-Cost Infection Prevention Clothing against Enveloped Viruses Such as SARS-CoV-2. ACS OMEGA 2021; 6:23495-23503. [PMID: 34514272 PMCID: PMC8424690 DOI: 10.1021/acsomega.1c03511] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/24/2021] [Indexed: 05/02/2023]
Abstract
Infection prevention clothing is becoming an essential protective tool in the current pandemic, especially because now we know that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can easily infect humans in poorly ventilated indoor spaces. However, commercial infection prevention clothing is made of fabrics that are not capable of inactivating the virus. Therefore, viral infections of symptomatic and asymptomatic individuals wearing protective clothing such as masks can occur through aerosol transmission or by contact with the contaminated surfaces of the masks, which are suspected as an increasing source of highly infectious biological waste. Herein, we report an easy fabrication method of a novel antiviral non-woven fabric containing polymer filaments that were coated with solidified hand soap. This extra protective fabric is capable of inactivating enveloped viruses such as SARS-CoV-2 and phage Φ6 within 1 min of contact. In this study, this antiviral fabric was used to fabricate an antiviral face mask and did not show any cytotoxic effect in human keratinocyte HaCaT cells. Furthermore, this antiviral non-woven fabric could be used for the fabrication of other infection prevention clothing such as caps, scrubs, shirts, trousers, disposable gowns, overalls, hoods, aprons, and shoe covers. Therefore, this low-cost technology could provide a wide range of infection-protective tools to combat COVID-19 and future pandemics in developed and underdeveloped countries.
Collapse
Affiliation(s)
- Alba Cano-Vicent
- Doctoral
School, Biomaterials and Bioengineering Laboratory, Centro de Investigación
Traslacional San Alberto Magno, Universidad
Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia 46001, Spain
| | - Alberto Tuñón-Molina
- Doctoral
School, Biomaterials and Bioengineering Laboratory, Centro de Investigación
Traslacional San Alberto Magno, Universidad
Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia 46001, Spain
| | - Miguel Martí
- Biomaterials
and Bioengineering Laboratory, Centro de Investigación Traslacional
San Alberto Magno, Universidad Católica
de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia 46001, Spain
| | - Yukiko Muramoto
- Laboratory
of Ultrastructural Virology, Institute for Frontier Life and Medical
Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Takeshi Noda
- Laboratory
of Ultrastructural Virology, Institute for Frontier Life and Medical
Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Kazuo Takayama
- Center
for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Ángel Serrano-Aroca
- Biomaterials
and Bioengineering Laboratory, Centro de Investigación Traslacional
San Alberto Magno, Universidad Católica
de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia 46001, Spain
| |
Collapse
|