Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Emerg Infect Dis ; 29(6): 1240-1243, 2023 06.
Article in English | MEDLINE | ID: covidwho-2315020

ABSTRACT

We performed 2 surveys during 2022 to estimate point prevalences of SARS-CoV-2 infection compared with overall seroprevalence in Sweden. Point prevalence was 1.4% in March and 1.5% in September. Estimated seroprevalence was >80%, including among unvaccinated children. Continued SARS-CoV-2 surveillance is necessary for detecting emerging, possibly more pathogenic variants.


Subject(s)
COVID-19 , Child , Humans , COVID-19/epidemiology , Prevalence , SARS-CoV-2 , Sweden/epidemiology , Seroepidemiologic Studies
3.
Cell Syst ; 13(8): 665-681.e4, 2022 Aug 17.
Article in English | MEDLINE | ID: covidwho-1982706

ABSTRACT

The clinical outcome and disease severity in coronavirus disease 2019 (COVID-19) are heterogeneous, and the progression or fatality of the disease cannot be explained by a single factor like age or comorbidities. In this study, we used system-wide network-based system biology analysis using whole blood RNA sequencing, immunophenotyping by flow cytometry, plasma metabolomics, and single-cell-type metabolomics of monocytes to identify the potential determinants of COVID-19 severity at personalized and group levels. Digital cell quantification and immunophenotyping of the mononuclear phagocytes indicated a substantial role in coordinating the immune cells that mediate COVID-19 severity. Stratum-specific and personalized genome-scale metabolic modeling indicated monocarboxylate transporter family genes (e.g., SLC16A6), nucleoside transporter genes (e.g., SLC29A1), and metabolites such as α-ketoglutarate, succinate, malate, and butyrate could play a crucial role in COVID-19 severity. Metabolic perturbations targeting the central metabolic pathway (TCA cycle) can be an alternate treatment strategy in severe COVID-19.


Subject(s)
COVID-19 , Humans , Metabolic Networks and Pathways , Metabolomics
4.
Mol Cell Proteomics ; 20: 100159, 2021.
Article in English | MEDLINE | ID: covidwho-1525879

ABSTRACT

Viruses hijack host metabolic pathways for their replicative advantage. In this study, using patient-derived multiomics data and in vitro infection assays, we aimed to understand the role of key metabolic pathways that can regulate severe acute respiratory syndrome coronavirus-2 reproduction and their association with disease severity. We used multiomics platforms (targeted and untargeted proteomics and untargeted metabolomics) on patient samples and cell-line models along with immune phenotyping of metabolite transporters in patient blood cells to understand viral-induced metabolic modulations. We also modulated key metabolic pathways that were identified using multiomics data to regulate the viral reproduction in vitro. Coronavirus disease 2019 disease severity was characterized by increased plasma glucose and mannose levels. Immune phenotyping identified altered expression patterns of carbohydrate transporter, glucose transporter 1, in CD8+ T cells, intermediate and nonclassical monocytes, and amino acid transporter, xCT, in classical, intermediate, and nonclassical monocytes. In in vitro lung epithelial cell (Calu-3) infection model, we found that glycolysis and glutaminolysis are essential for virus replication, and blocking these metabolic pathways caused significant reduction in virus production. Taken together, we therefore hypothesized that severe acute respiratory syndrome coronavirus-2 utilizes and rewires pathways governing central carbon metabolism leading to the efflux of toxic metabolites and associated with disease severity. Thus, the host metabolic perturbation could be an attractive strategy to limit the viral replication and disease severity.


Subject(s)
Blood Proteins/metabolism , COVID-19/etiology , SARS-CoV-2/physiology , Adult , Aged , Amino Acid Transport System y+/blood , Amino Acids/blood , Biomarkers/blood , Blood Proteins/analysis , COVID-19/metabolism , COVID-19/virology , Carbohydrates/blood , Case-Control Studies , Glucose Transporter Type 1/blood , Hospitalization , Humans , Immunophenotyping , Mannose/blood , Mannose-Binding Lectin/blood , Middle Aged , Severity of Illness Index , Virus Replication
5.
J Proteome Res ; 19(11): 4259-4274, 2020 11 06.
Article in English | MEDLINE | ID: covidwho-960274

ABSTRACT

Emerging and re-emerging infectious diseases due to RNA viruses cause major negative consequences for the quality of life, public health, and overall economic development. Most of the RNA viruses causing illnesses in humans are of zoonotic origin. Zoonotic viruses can directly be transferred from animals to humans through adaptation, followed by human-to-human transmission, such as in human immunodeficiency virus (HIV), severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and, more recently, SARS coronavirus 2 (SARS-CoV-2), or they can be transferred through insects or vectors, as in the case of Crimean-Congo hemorrhagic fever virus (CCHFV), Zika virus (ZIKV), and dengue virus (DENV). At the present, there are no vaccines or antiviral compounds against most of these viruses. Because proteins possess a vast array of functions in all known biological systems, proteomics-based strategies can provide important insights into the investigation of disease pathogenesis and the identification of promising antiviral drug targets during an epidemic or pandemic. Mass spectrometry technology has provided the capacity required for the precise identification and the sensitive and high-throughput analysis of proteins on a large scale and has contributed greatly to unravelling key protein-protein interactions, discovering signaling networks, and understanding disease mechanisms. In this Review, we present an account of quantitative proteomics and its application in some prominent recent examples of emerging and re-emerging RNA virus diseases like HIV-1, CCHFV, ZIKV, and DENV, with more detail with respect to coronaviruses (MERS-CoV and SARS-CoV) as well as the recent SARS-CoV-2 pandemic.


Subject(s)
Communicable Diseases, Emerging , Proteomics , RNA Virus Infections , Animals , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Communicable Diseases, Emerging/diagnosis , Communicable Diseases, Emerging/therapy , Communicable Diseases, Emerging/virology , Coronavirus Infections/diagnosis , Humans , Pandemics , Pneumonia, Viral , RNA Virus Infections/diagnosis , RNA Virus Infections/therapy , RNA Virus Infections/virology , RNA Viruses
6.
Emerg Microbes Infect ; 9(1): 1748-1760, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-658315

ABSTRACT

How severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections engage cellular host pathways and innate immunity in infected cells remains largely elusive. We performed an integrative proteo-transcriptomics analysis in SARS-CoV-2 infected Huh7 cells to map the cellular response to the invading virus over time. We identified four pathways, ErbB, HIF-1, mTOR and TNF signaling, among others that were markedly modulated during the course of the SARS-CoV-2 infection in vitro. Western blot validation of the downstream effector molecules of these pathways revealed a dose-dependent activation of Akt, mTOR, S6K1 and 4E-BP1 at 24 hours post infection (hpi). However, we found a significant inhibition of HIF-1α through 24hpi and 48hpi of the infection, suggesting a crosstalk between the SARS-CoV-2 and the Akt/mTOR/HIF-1 signaling pathways. Inhibition of the mTOR signaling pathway using Akt inhibitor MK-2206 showed a significant reduction in virus production. Further investigations are required to better understand the molecular sequelae in order to guide potential therapy in the management of severe coronavirus disease 2019 (COVID-19) patients.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/virology , Gene Expression Profiling/methods , Pneumonia, Viral/virology , Proteomics/methods , Signal Transduction , COVID-19 , Cell Line , Chromatography, Liquid , Coronavirus Infections/genetics , Coronavirus Infections/metabolism , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Pandemics , Pneumonia, Viral/genetics , Pneumonia, Viral/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , SARS-CoV-2 , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL