Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
IISE Annual Conference and Expo 2022 ; 2022.
Article in English | Scopus | ID: covidwho-2011555

ABSTRACT

Recent events such as natural hazards, diet trends, and the COVID-19 pandemic have shed light on several inefficiencies of the traditional fresh fruit and vegetable (FFV) supply chain (SC). Factors that contribute to this problem are the lack of coordination of the SC participants, the inaccessibility of planning tools for agricultural production, and the absence of market information to determine if a product will have a demand. Intelligent SCs are emerging to address some of these issues by using data-driven tools to aid in decision-making. Nonetheless, there has been little work to incorporate market intelligence in the new SC model to solve the lack of market information in the traditional model. It is essential to include market intelligence in the new SC model to decrease food waste, reduce losses related to low market prices and demands, and avoid scarcity events in which food availability and affordability decrease, while aiding small growers by alerting them of potential market opportunities. This work aims to develop a market intelligence framework for the FFV SC and incorporate it into the intelligent SC. A layered system approach is used with the goal of collecting relevant data to monitor and diagnose the market's state and provide recommendations to the SC participants. The layered system framework aims to decompose the overall problem into several layers with distinct goals such as data collection, processing, monitoring, diagnostics, among others. This work will focus on the monitoring aspect of the system. © 2022 IISE Annual Conference and Expo 2022. All rights reserved.

2.
J Am Coll Emerg Physicians Open ; 1(4): 569-577, 2020 Aug.
Article in English | MEDLINE | ID: covidwho-1898677

ABSTRACT

Background: The SARS-CoV-2 (COVID-19) virus has wide community spread. The aim of this study was to describe patient characteristics and to identify factors associated with COVID-19 among emergency department (ED) patients under investigation for COVID-19 who were admitted to the hospital. Methods: This was a retrospective observational study from 8 EDs within a 9-hospital health system. Patients with COVID-19 testing around the time of hospital admission were included. The primary outcome measure was COVID-19 test result. Patient characteristics were described and a multivariable logistic regression model was used to identify factors associated with a positive COVID-19 test. Results: During the study period from March 1, 2020 to April 8, 2020, 2182 admitted patients had a test resulted for COVID-19. Of these patients, 786 (36%) had a positive test result. For COVID-19-positive patients, 63 (8.1%) died during hospitalization. COVID-19-positive patients had lower pulse oximetry (0.91 [95% confidence interval, CI], [0.88-0.94]), higher temperatures (1.36 [1.26-1.47]), and lower leukocyte counts than negative patients (0.78 [0.75-0.82]). Chronic lung disease (odds ratio [OR] 0.68, [0.52-0.90]) and histories of alcohol (0.64 [0.42-0.99]) or substance abuse (0.39 [0.25-0.62]) were less likely to be associated with a positive COVID-19 result. Conclusion: We observed a high percentage of positive results among an admitted ED cohort under investigation for COVID-19. Patient factors may be useful in early differentiation of patients with COVID-19 from similarly presenting respiratory illnesses although no single factor will serve this purpose.

3.
Nat Commun ; 13(1): 440, 2022 01 21.
Article in English | MEDLINE | ID: covidwho-1641960

ABSTRACT

Dysregulated immune responses against the SARS-CoV-2 virus are instrumental in severe COVID-19. However, the immune signatures associated with immunopathology are poorly understood. Here we use multi-omics single-cell analysis to probe the dynamic immune responses in hospitalized patients with stable or progressive course of COVID-19, explore V(D)J repertoires, and assess the cellular effects of tocilizumab. Coordinated profiling of gene expression and cell lineage protein markers shows that S100Ahi/HLA-DRlo classical monocytes and activated LAG-3hi T cells are hallmarks of progressive disease and highlights the abnormal MHC-II/LAG-3 interaction on myeloid and T cells, respectively. We also find skewed T cell receptor repertories in expanded effector CD8+ clones, unmutated IGHG+ B cell clones, and mutated B cell clones with stable somatic hypermutation frequency over time. In conclusion, our in-depth immune profiling reveals dyssynchrony of the innate and adaptive immune interaction in progressive COVID-19.


Subject(s)
Adaptive Immunity/immunology , COVID-19/immunology , Gene Expression Profiling/methods , Immunity, Innate/immunology , SARS-CoV-2/immunology , Single-Cell Analysis/methods , Adaptive Immunity/drug effects , Adaptive Immunity/genetics , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , COVID-19/genetics , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Male , RNA-Seq/methods , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , COVID-19 Drug Treatment
4.
Sci Rep ; 11(1): 11049, 2021 05 26.
Article in English | MEDLINE | ID: covidwho-1246386

ABSTRACT

The SARS-CoV-2 pandemic has challenged researchers at a global scale. The scientific community's massive response has resulted in a flood of experiments, analyses, hypotheses, and publications, especially in the field of drug repurposing. However, many of the proposed therapeutic compounds obtained from SARS-CoV-2 specific assays are not in agreement and thus demonstrate the need for a singular source of COVID-19 related information from which a rational selection of drug repurposing candidates can be made. In this paper, we present the COVID-19 PHARMACOME, a comprehensive drug-target-mechanism graph generated from a compilation of 10 separate disease maps and sources of experimental data focused on SARS-CoV-2/COVID-19 pathophysiology. By applying our systematic approach, we were able to predict the synergistic effect of specific drug pairs, such as Remdesivir and Thioguanosine or Nelfinavir and Raloxifene, on SARS-CoV-2 infection. Experimental validation of our results demonstrate that our graph can be used to not only explore the involved mechanistic pathways, but also to identify novel combinations of drug repurposing candidates.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Drug Repositioning/methods , SARS-CoV-2/physiology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/therapeutic use , Alanine/analogs & derivatives , Alanine/therapeutic use , Combined Modality Therapy , Computational Biology , Drug Synergism , Drug Therapy, Combination , GTP Phosphohydrolases/therapeutic use , Humans , Knowledge Bases , Nelfinavir/therapeutic use , Pandemics , Raloxifene Hydrochloride/therapeutic use
5.
Diabetic Medicine ; 38(SUPPL 1):59, 2021.
Article in English | EMBASE | ID: covidwho-1238394

ABSTRACT

Aims: Historically the National Diabetes Prevention Programme (NDPP) has low retention rates which are accentuated in the BAME population. We aimed to design and deliver a fun, alternative type 2 diabetes prevention programme that would have an increased retention rate and increase participant's physical activity levels. The Diabetes Prevention Decathlon (DPD) structured education programme featured;weekly health and well-being workshops and physical activity sessions, gamification and the use of a mobile phone application that incentivised continued physical activity between sessions. Method: An innovation grant awarded the opportunity for collaboration between multiple partners within the NHS, public health, community charities, patient groups, and a digital enterprise. This resulted in the creation of the DPD which was piloted in a deprived South West London borough. The inclusion criteria for participants included a HbA1c between 42 and 47 mmol/mol (6.0-6.4%), indicative for non-diabetic hypoglycaemia. Referrals, retention rates, weight loss and physical activity data were recorded over the ten-week programme alongside key demographic markers. Results: With an 87% completion rate (n31), 39 kg group weight loss and 40% increase in activity levels, the programme yielded excellent results compared to the NDPP. Semi-structured interviews highlighted the importance of group interactions and regular opportunities to be physically active. Positive but unplanned outcomes were the high BAME uptake of 77% and subsequent retention 67%, and covid driven agile conversion to virtual delivery. Conclusions: The DPD successfully piloted a new and fun approach to diabetes prevention with further intentions to work with local communities to increase and improve BAME resources.

6.
Immunity ; 54(5): 1083-1095.e7, 2021 05 11.
Article in English | MEDLINE | ID: covidwho-1179682

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a life-threatening post-infectious complication occurring unpredictably weeks after mild or asymptomatic SARS-CoV-2 infection. We profiled MIS-C, adult COVID-19, and healthy pediatric and adult individuals using single-cell RNA sequencing, flow cytometry, antigen receptor repertoire analysis, and unbiased serum proteomics, which collectively identified a signature in MIS-C patients that correlated with disease severity. Despite having no evidence of active infection, MIS-C patients had elevated S100A-family alarmins and decreased antigen presentation signatures, indicative of myeloid dysfunction. MIS-C patients showed elevated expression of cytotoxicity genes in NK and CD8+ T cells and expansion of specific IgG-expressing plasmablasts. Clinically severe MIS-C patients displayed skewed memory T cell TCR repertoires and autoimmunity characterized by endothelium-reactive IgG. The alarmin, cytotoxicity, TCR repertoire, and plasmablast signatures we defined have potential for application in the clinic to better diagnose and potentially predict disease severity early in the course of MIS-C.


Subject(s)
COVID-19/immunology , COVID-19/pathology , SARS-CoV-2/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Adolescent , Alarmins/immunology , Autoantibodies/immunology , CD8-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Cytotoxicity, Immunologic/genetics , Endothelium/immunology , Endothelium/pathology , Humans , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Plasma Cells/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Severity of Illness Index
7.
PLoS Biol ; 19(3): e3001143, 2021 03.
Article in English | MEDLINE | ID: covidwho-1138557

ABSTRACT

There are currently limited Food and Drug Administration (FDA)-approved drugs and vaccines for the treatment or prevention of Coronavirus Disease 2019 (COVID-19). Enhanced understanding of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and pathogenesis is critical for the development of therapeutics. To provide insight into viral replication, cell tropism, and host-viral interactions of SARS-CoV-2, we performed single-cell (sc) RNA sequencing (RNA-seq) of experimentally infected human bronchial epithelial cells (HBECs) in air-liquid interface (ALI) cultures over a time course. This revealed novel polyadenylated viral transcripts and highlighted ciliated cells as a major target at the onset of infection, which we confirmed by electron and immunofluorescence microscopy. Over the course of infection, the cell tropism of SARS-CoV-2 expands to other epithelial cell types including basal and club cells. Infection induces cell-intrinsic expression of type I and type III interferons (IFNs) and interleukin (IL)-6 but not IL-1. This results in expression of interferon-stimulated genes (ISGs) in both infected and bystander cells. This provides a detailed characterization of genes, cell types, and cell state changes associated with SARS-CoV-2 infection in the human airway.


Subject(s)
Bronchi/pathology , COVID-19/diagnosis , Gene Expression , SARS-CoV-2/isolation & purification , Single-Cell Analysis/methods , Adult , Bronchi/virology , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Cells, Cultured , Epithelium/pathology , Epithelium/virology , Humans , Immunity, Innate , Longitudinal Studies , SARS-CoV-2/genetics , Transcriptome , Viral Tropism
8.
Cell ; 184(1): 76-91.e13, 2021 01 07.
Article in English | MEDLINE | ID: covidwho-1064906

ABSTRACT

Identification of host genes essential for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may reveal novel therapeutic targets and inform our understanding of coronavirus disease 2019 (COVID-19) pathogenesis. Here we performed genome-wide CRISPR screens in Vero-E6 cells with SARS-CoV-2, Middle East respiratory syndrome CoV (MERS-CoV), bat CoV HKU5 expressing the SARS-CoV-1 spike, and vesicular stomatitis virus (VSV) expressing the SARS-CoV-2 spike. We identified known SARS-CoV-2 host factors, including the receptor ACE2 and protease Cathepsin L. We additionally discovered pro-viral genes and pathways, including HMGB1 and the SWI/SNF chromatin remodeling complex, that are SARS lineage and pan-coronavirus specific, respectively. We show that HMGB1 regulates ACE2 expression and is critical for entry of SARS-CoV-2, SARS-CoV-1, and NL63. We also show that small-molecule antagonists of identified gene products inhibited SARS-CoV-2 infection in monkey and human cells, demonstrating the conserved role of these genetic hits across species. This identifies potential therapeutic targets for SARS-CoV-2 and reveals SARS lineage-specific and pan-CoV host factors that regulate susceptibility to highly pathogenic CoVs.


Subject(s)
Coronavirus Infections/genetics , Genome-Wide Association Study , Host-Pathogen Interactions , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/immunology , COVID-19/virology , Cell Line , Chlorocebus aethiops , Clustered Regularly Interspaced Short Palindromic Repeats , Coronavirus/classification , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Gene Knockout Techniques , Gene Regulatory Networks , HEK293 Cells , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Host-Pathogen Interactions/drug effects , Humans , Vero Cells , Virus Internalization
9.
J Exp Med ; 218(3)2021 03 01.
Article in English | MEDLINE | ID: covidwho-1024074

ABSTRACT

Although COVID-19 is considered to be primarily a respiratory disease, SARS-CoV-2 affects multiple organ systems including the central nervous system (CNS). Yet, there is no consensus on the consequences of CNS infections. Here, we used three independent approaches to probe the capacity of SARS-CoV-2 to infect the brain. First, using human brain organoids, we observed clear evidence of infection with accompanying metabolic changes in infected and neighboring neurons. However, no evidence for type I interferon responses was detected. We demonstrate that neuronal infection can be prevented by blocking ACE2 with antibodies or by administering cerebrospinal fluid from a COVID-19 patient. Second, using mice overexpressing human ACE2, we demonstrate SARS-CoV-2 neuroinvasion in vivo. Finally, in autopsies from patients who died of COVID-19, we detect SARS-CoV-2 in cortical neurons and note pathological features associated with infection with minimal immune cell infiltrates. These results provide evidence for the neuroinvasive capacity of SARS-CoV-2 and an unexpected consequence of direct infection of neurons by SARS-CoV-2.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antibodies, Blocking/chemistry , COVID-19 , Cerebral Cortex , Neurons , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/metabolism , COVID-19/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/virology , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Neurons/metabolism , Neurons/pathology , Neurons/virology , Organoids/metabolism , Organoids/pathology , Organoids/virology
10.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.12.01.20241364

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a life-threatening post-infectious complication occurring unpredictably weeks after mild or asymptomatic SARS-CoV2 infection in otherwise healthy children. Here, we define immune abnormalities in MIS-C compared to adult COVID-19 and pediatric/adult healthy controls using single-cell RNA sequencing, antigen receptor repertoire analysis, unbiased serum proteomics, and in vitro assays. Despite no evidence of active infection, we uncover elevated S100A-family alarmins in myeloid cells and marked enrichment of serum proteins that map to myeloid cells and pathways including cytokines, complement/coagulation, and fluid shear stress in MIS-C patients. Moreover, NK and CD8 T cell cytotoxicity genes are elevated, and plasmablasts harboring IgG1 and IgG3 are expanded. Consistently, we detect elevated binding of serum IgG from severe MIS-C patients to activated human cardiac microvascular endothelial cells in culture. Thus, we define immunopathology features of MIS-C with implications for predicting and managing this SARS-CoV2-induced critical illness in children.


Subject(s)
Cryopyrin-Associated Periodic Syndromes , Severe Acute Respiratory Syndrome , Critical Illness , Drug-Related Side Effects and Adverse Reactions , COVID-19
11.
Ann Emerg Med ; 76(4): 442-453, 2020 10.
Article in English | MEDLINE | ID: covidwho-813459

ABSTRACT

STUDY OBJECTIVE: The goal of this study is to create a predictive, interpretable model of early hospital respiratory failure among emergency department (ED) patients admitted with coronavirus disease 2019 (COVID-19). METHODS: This was an observational, retrospective, cohort study from a 9-ED health system of admitted adult patients with severe acute respiratory syndrome coronavirus 2 (COVID-19) and an oxygen requirement less than or equal to 6 L/min. We sought to predict respiratory failure within 24 hours of admission as defined by oxygen requirement of greater than 10 L/min by low-flow device, high-flow device, noninvasive or invasive ventilation, or death. Predictive models were compared with the Elixhauser Comorbidity Index, quick Sequential [Sepsis-related] Organ Failure Assessment, and the CURB-65 pneumonia severity score. RESULTS: During the study period, from March 1 to April 27, 2020, 1,792 patients were admitted with COVID-19, 620 (35%) of whom had respiratory failure in the ED. Of the remaining 1,172 admitted patients, 144 (12.3%) met the composite endpoint within the first 24 hours of hospitalization. On the independent test cohort, both a novel bedside scoring system, the quick COVID-19 Severity Index (area under receiver operating characteristic curve mean 0.81 [95% confidence interval {CI} 0.73 to 0.89]), and a machine-learning model, the COVID-19 Severity Index (mean 0.76 [95% CI 0.65 to 0.86]), outperformed the Elixhauser mortality index (mean 0.61 [95% CI 0.51 to 0.70]), CURB-65 (0.50 [95% CI 0.40 to 0.60]), and quick Sequential [Sepsis-related] Organ Failure Assessment (0.59 [95% CI 0.50 to 0.68]). A low quick COVID-19 Severity Index score was associated with a less than 5% risk of respiratory decompensation in the validation cohort. CONCLUSION: A significant proportion of admitted COVID-19 patients progress to respiratory failure within 24 hours of admission. These events are accurately predicted with bedside respiratory examination findings within a simple scoring system.


Subject(s)
Coronavirus Infections/complications , Coronavirus Infections/diagnosis , Emergency Service, Hospital , Pneumonia, Viral/complications , Pneumonia, Viral/diagnosis , Respiratory Insufficiency/virology , Severity of Illness Index , Adolescent , Adult , Aged , Betacoronavirus , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Coronavirus Infections/therapy , Female , Humans , Male , Middle Aged , Oxygen Inhalation Therapy , Pandemics , Pneumonia, Viral/therapy , Respiratory Insufficiency/therapy , Retrospective Studies , Risk Assessment/methods , SARS-CoV-2 , Young Adult
12.
PLoS One ; 15(9): e0238829, 2020.
Article in English | MEDLINE | ID: covidwho-807468

ABSTRACT

BACKGROUND: Patients with comorbid conditions have a higher risk of mortality with SARS-CoV-2 (COVID-19) infection, but the impact on heart failure patients living near a disease hotspot is unknown. Therefore, we sought to characterize the prevalence and outcomes of COVID-19 in a live registry of heart failure patients across an integrated health care system in Connecticut. METHODS: In this retrospective analysis, the Yale Heart Failure Registry (NCT04237701) that includes 26,703 patients with heart failure across a 6-hospital integrated health care system in Connecticut was queried on April 16th, 2020 for all patients tested for COVID-19. Sociodemographic and geospatial data as well as, clinical management, respiratory failure, and patient mortality were obtained via the real-time registry. Data on COVID-19 specific care was extracted by retrospective chart review. RESULTS: COVID-19 testing was performed on 900 symptomatic patients, comprising 3.4% of the Yale Heart Failure Registry (N = 26,703). Overall, 206 (23%) were COVID- 19+. As compared to COVID-19-, these patients were more likely to be older, black, have hypertension, coronary artery disease, and were less likely to be on renin angiotensin blockers (P<0.05, all). COVID-19- patients tended to be more diffusely spread across the state whereas COVID-19+ were largely clustered around urban centers. 20% of COVID-19+ patients died, and age was associated with increased risk of death [OR 1.92 95% CI (1.33-2.78); P<0.001]. Among COVID-19+ patients who were ≥85 years of age rates of hospitalization were 87%, rates of death 36%, and continuing hospitalization 62% at time of manuscript preparation. CONCLUSIONS: In this real-world snapshot of COVID-19 infection among a large cohort of heart failure patients, we found that a small proportion had undergone testing. Patients found to be COVID-19+ tended to be black with multiple comorbidities and clustered around lower socioeconomic status communities. Elderly COVID-19+ patients were very likely to be admitted to the hospital and experience high rates of mortality.


Subject(s)
Coronavirus Infections/epidemiology , Coronavirus Infections/mortality , Heart Failure/epidemiology , Pneumonia, Viral/epidemiology , Pneumonia, Viral/mortality , Registries , Aged , Aged, 80 and over , Betacoronavirus , COVID-19 , Cohort Studies , Comorbidity , Connecticut , Delivery of Health Care, Integrated , Female , Heart Failure/mortality , Hospitalization/statistics & numerical data , Humans , Male , Middle Aged , Pandemics , Retrospective Studies , SARS-CoV-2
13.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.07.16.20153437

ABSTRACT

A dysregulated immune response against the SARS-CoV-2 virus plays a critical role in severe COVID-19. However, the molecular and cellular mechanisms by which the virus causes lethal immunopathology are poorly understood. Here, we utilize multi-omics single-cell analysis to probe dynamic immune responses in patients with stable or progressive manifestations of COVID-19, and assess the effects of tocilizumab, an anti-IL-6 receptor monoclonal antibody. Coordinated profiling of gene expression and cell lineage protein markers reveals a prominent type-1 interferon response across all immune cells, especially in progressive patients. An anti-inflammatory innate immune response and a pre-exhaustion phenotype in activated T cells are hallmarks of progressive disease. Skewed T cell receptor repertoires in CD8 T cells and uniquely enriched V(D)J sequences are also identified in COVID-19 patients. B cell repertoire and somatic hypermutation analysis are consistent with a primary immune response, with possible contribution from memory B cells. Our in-depth immune profiling reveals dyssynchrony of the innate and adaptive immune interaction in progressive COVID-19, which may contribute to delayed virus clearance and has implications for therapeutic intervention.


Subject(s)
COVID-19
14.
arxiv; 2020.
Preprint in English | PREPRINT-ARXIV | ID: ppzbmed-2006.12971v2

ABSTRACT

A molecular and cellular understanding of how SARS-CoV-2 variably infects and causes severe COVID-19 remains a bottleneck in developing interventions to end the pandemic. We sought to use deep learning to study the biology of SARS-CoV-2 infection and COVID-19 severity by identifying transcriptomic patterns and cell types associated with SARS-CoV-2 infection and COVID-19 severity. To do this, we developed a new approach to generating self-supervised edge features. We propose a model that builds on Graph Attention Networks (GAT), creates edge features using self-supervised learning, and ingests these edge features via a Set Transformer. This model achieves significant improvements in predicting the disease state of individual cells, given their transcriptome. We apply our model to single-cell RNA sequencing datasets of SARS-CoV-2 infected lung organoids and bronchoalveolar lavage fluid samples of patients with COVID-19, achieving state-of-the-art performance on both datasets with our model. We then borrow from the field of explainable AI (XAI) to identify the features (genes) and cell types that discriminate bystander vs. infected cells across time and moderate vs. severe COVID-19 disease. To the best of our knowledge, this represents the first application of deep learning to identifying the molecular and cellular determinants of SARS-CoV-2 infection and COVID-19 severity using single-cell omics data.


Subject(s)
COVID-19 , Severe Acute Respiratory Syndrome , Lung Diseases
15.
arxiv; 2020.
Preprint in English | PREPRINT-ARXIV | ID: ppzbmed-2007.04777v1

ABSTRACT

Graph Neural Networks (GNN) have been extensively used to extract meaningful representations from graph structured data and to perform predictive tasks such as node classification and link prediction. In recent years, there has been a lot of work incorporating edge features along with node features for prediction tasks. One of the main difficulties in using edge features is that they are often handcrafted, hard to get, specific to a particular domain, and may contain redundant information. In this work, we present a framework for creating new edge features, applicable to any domain, via a combination of self-supervised and unsupervised learning. In addition to this, we use Forman-Ricci curvature as an additional edge feature to encapsulate the local geometry of the graph. We then encode our edge features via a Set Transformer and combine them with node features extracted from popular GNN architectures for node classification in an end-to-end training scheme. We validate our work on three biological datasets comprising of single-cell RNA sequencing data of neurological disease, \textit{in vitro} SARS-CoV-2 infection, and human COVID-19 patients. We demonstrate that our method achieves better performance on node classification tasks over baseline Graph Attention Network (GAT) and Graph Convolutional Network (GCN) models. Furthermore, given the attention mechanism on edge and node features, we are able to interpret the cell types and genes that determine the course and severity of COVID-19, contributing to a growing list of potential disease biomarkers and therapeutic targets.


Subject(s)
COVID-19 , Heredodegenerative Disorders, Nervous System
16.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.05.07.20094573

ABSTRACT

ObjectiveThe goal of this study was to create a predictive model of early hospital respiratory decompensation among patients with COVID-19. DesignObservational, retrospective cohort study. SettingNine-hospital health system within the Northeastern United States. PopulationsAdult patients ([≥] 18 years) admitted from the emergency department who tested positive for SARS-CoV-2 (COVID-19) up to 24 hours after initial presentation. Patients meeting criteria for respiratory critical illness within 4 hours of arrival were excluded. Main outcome and performance measuresWe used a composite endpoint of critical illness as defined by oxygen requirement (greater than 10 L/min by low-flow device, high-flow device, non-invasive, or invasive ventilation) or death within the first 24 hours of hospitalization. We developed models predicting our composite endpoint using patient demographic and clinical data available within the first four hours of arrival. Eight hospitals (n = 932) were used for model development and one hospital (n = 240) was held out for external validation. Area under receiver operating characteristic (AU-ROC), precision-recall curves (AU-PRC), and calibration metrics were used to compare predictive models to three illness scoring systems: Elixhauser comorbidity index, qSOFA, and CURB-65. ResultsDuring the study period from March 1, 2020 to April 27,2020, 1,792 patients were admitted with COVID-19. Six-hundred and twenty patients were excluded based on age or critical illness within the first 4 hours, yielding 1,172 patients in the final cohort. Of these patients, 144 (12.3%) met the composite endpoint within the first 24 hours. We first developed a bedside quick COVID-19 severity index (qCSI), a twelve-point scale using nasal cannula flow rate, respiratory rate, and minimum documented pulse oximetry. We then created a machine-learning gradient boosting model, the COVID-19 severity index (CSI), using twelve additional variables including inflammatory markers and liver chemistries. Both the qCSI (AU-ROC mean [95% CI]: 0.90 [0.85-0.96]) and CSI (AU-ROC: 0.91 [0.86-0.97]) outperformed the comparator models (qSOFA: 0.76 [0.69-0.85]; Elixhauser: 0.70 [0.62-0.80]; CURB-65: AU-ROC 0.66 [0.58-0.77]) on cross-validation and performed well on external validation (qCSI: 0.82, CSI: 0.76, CURB-65: 0.50, qSOFA: 0.59, Elixhauser: 0.61). We find that a qCSI score of 0-3 is associated with a less than 5% risk of critical respiratory illness, while a score of 9-12 is associated with a 57% risk of progression to critical illness. ConclusionsA significant proportion of admitted COVID-19 patients decompensate within 24 hours of hospital presentation and these events are accurately predicted using bedside respiratory exam findings within a simple scoring system.


Subject(s)
COVID-19
17.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.05.06.081695

ABSTRACT

SARS-CoV-2, the causative agent of COVID-19, has tragically burdened individuals and institutions around the world. There are currently no approved drugs or vaccines for the treatment or prevention of COVID-19. Enhanced understanding of SARS-CoV-2 infection and pathogenesis is critical for the development of therapeutics. To reveal insight into viral replication, cell tropism, and host-viral interactions of SARS-CoV-2 we performed single-cell RNA sequencing of experimentally infected human bronchial epithelial cells (HBECs) in air-liquid interface cultures over a time-course. This revealed novel polyadenylated viral transcripts and highlighted ciliated cells as a major target of infection, which we confirmed by electron microscopy. Over the course of infection, cell tropism of SARS-CoV-2 expands to other epithelial cell types including basal and club cells. Infection induces cell-intrinsic expression of type I and type III IFNs and IL6 but not IL1. This results in expression of interferon-stimulated genes in both infected and bystander cells. We observe similar gene expression changes from a COVID-19 patient ex vivo. In addition, we developed a new computational method termed CONditional DENSity Embedding (CONDENSE) to characterize and compare temporal gene dynamics in response to infection, which revealed genes relating to endothelin, angiogenesis, interferon, and inflammation-causing signaling pathways. In this study, we conducted an in-depth analysis of SARS-CoV-2 infection in HBECs and a COVID-19 patient and revealed genes, cell types, and cell state changes associated with infection.


Subject(s)
COVID-19 , Inflammation
SELECTION OF CITATIONS
SEARCH DETAIL