Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Proc Natl Acad Sci U S A ; 120(51): e2300474120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38100417

ABSTRACT

Seasonal influenza results in 3 to 5 million cases of severe disease and 250,000 to 500,000 deaths annually. Macrophages have been implicated in both the resolution and progression of the disease, but the drivers of these outcomes are poorly understood. We probed mouse lung transcriptomic datasets using the Digital Cell Quantifier algorithm to predict immune cell subsets that correlated with mild or severe influenza A virus (IAV) infection outcomes. We identified a unique lung macrophage population that transcriptionally resembled small serosal cavity macrophages and whose presence correlated with mild disease. Until now, the study of serosal macrophage translocation in the context of viral infections has been neglected. Here, we show that pleural macrophages (PMs) migrate from the pleural cavity to the lung after infection with IAV. We found that the depletion of PMs increased morbidity and pulmonary inflammation. There were increased proinflammatory cytokines in the pleural cavity and an influx of neutrophils within the lung. Our results show that PMs are recruited to the lung during IAV infection and contribute to recovery from influenza. This study expands our knowledge of PM plasticity and identifies a source of lung macrophages independent of monocyte recruitment and local proliferation.


Subject(s)
Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Animals , Mice , Humans , Influenza, Human/genetics , Lung , Macrophages , Macrophages, Alveolar
2.
Cell Syst ; 13(12): 1002-1015.e9, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36516834

ABSTRACT

When challenged with an invading pathogen, the host-defense response is engaged to eliminate the pathogen (resistance) and to maintain health in the presence of the pathogen (disease tolerance). However, the identification of distinct molecular programs underpinning disease tolerance and resistance remained obscure. We exploited transcriptional and physiological monitoring across 33 mouse strains, during in vivo influenza virus infection, to identify two host-defense gene programs-one is associated with hallmarks of disease tolerance and the other with hallmarks of resistance. Both programs constitute generic responses in multiple mouse and human cell types. Our study describes the organizational principles of these programs and validates Arhgdia as a regulator of disease-tolerance states in epithelial cells. We further reveal that the baseline disease-tolerance state in peritoneal macrophages is associated with the pathophysiological response to injury and infection. Our framework provides a paradigm for the understanding of disease tolerance and resistance at the molecular level.


Subject(s)
Influenza, Human , Orthomyxoviridae Infections , Mice , Humans , Animals , Influenza, Human/genetics , Host-Pathogen Interactions/genetics , Orthomyxoviridae Infections/genetics , Epithelial Cells/metabolism
3.
Genetics ; 217(4)2021 04 15.
Article in English | MEDLINE | ID: mdl-33734353

ABSTRACT

Recent computational methods have enabled the inference of the cell-type-specificity of eQTLs based on bulk transcriptomes from highly heterogeneous tissues. However, these methods are limited in their scalability to highly heterogeneous tissues and limited in their broad applicability to any cell-type specificity of eQTLs. Here we present and demonstrate Cell Lineage Genetics (CeL-Gen), a novel computational approach that allows inference of eQTLs together with the subsets of cell types in which they have an effect, from bulk transcriptome data. To obtain improved scalability and broader applicability, CeL-Gen takes as input the known cell lineage tree and relies on the observation that dynamic changes in genetic effects occur relatively infrequently during cell differentiation. CeL-Gen can therefore be used not only to tease apart genetic effects derived from different cell types but also to infer the particular differentiation steps in which genetic effects are altered.


Subject(s)
Cell Lineage , Genetic Variation , Genome-Wide Association Study/methods , Animals , Cell Differentiation , Humans , Quantitative Trait Loci , Transcriptome
4.
J Med Internet Res ; 22(10): e23197, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32961527

ABSTRACT

BACKGROUND: Patient-facing digital health tools have been promoted to help patients manage concerns related to COVID-19 and to enable remote care and self-care during the COVID-19 pandemic. It has also been suggested that these tools can help further our understanding of the clinical characteristics of this new disease. However, there is limited information on the characteristics and use patterns of these tools in practice. OBJECTIVE: The aims of this study are to describe the characteristics of people who use digital health tools to address COVID-19-related concerns; explore their self-reported symptoms and characterize the association of these symptoms with COVID-19; and characterize the recommendations provided by digital health tools. METHODS: This study used data from three digital health tools on the K Health app: a protocol-based COVID-19 self-assessment, an artificial intelligence (AI)-driven symptom checker, and communication with remote physicians. Deidentified data were extracted on the demographic and clinical characteristics of adults seeking COVID-19-related health information between April 8 and June 20, 2020. Analyses included exploring features associated with COVID-19 positivity and features associated with the choice to communicate with a remote physician. RESULTS: During the period assessed, 71,619 individuals completed the COVID-19 self-assessment, 41,425 also used the AI-driven symptom checker, and 2523 consulted with remote physicians. Individuals who used the COVID-19 self-assessment were predominantly female (51,845/71,619, 72.4%), with a mean age of 34.5 years (SD 13.9). Testing for COVID-19 was reported by 2901 users, of whom 433 (14.9%) reported testing positive. Users who tested positive for COVID-19 were more likely to have reported loss of smell or taste (relative rate [RR] 6.66, 95% CI 5.53-7.94) and other established COVID-19 symptoms as well as ocular symptoms. Users communicating with a remote physician were more likely to have been recommended by the self-assessment to undergo immediate medical evaluation due to the presence of severe symptoms (RR 1.19, 95% CI 1.02-1.32). Most consultations with remote physicians (1940/2523, 76.9%) were resolved without need for referral to an in-person visit or to the emergency department. CONCLUSIONS: Our results suggest that digital health tools can help support remote care and self-management of COVID-19 and that self-reported symptoms from digital interactions can extend our understanding of the symptoms associated with COVID-19.


Subject(s)
Clinical Laboratory Techniques , Coronavirus Infections/diagnosis , Pneumonia, Viral/diagnosis , Adult , Artificial Intelligence , Betacoronavirus , COVID-19 , COVID-19 Testing , Female , Humans , Male , Pandemics , Referral and Consultation , Retrospective Studies , SARS-CoV-2 , Self Report
5.
Transl Lung Cancer Res ; 9(3): 682-692, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32676330

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is the most common cause of cancer-death due to early metastatic spread, in many cases primarily to the brain. Organ-specific pattern of spread of disease might be driven by the activity of a specific signaling pathway within the primary tumors. We aimed to identify an expression signature of genes and the relevant signaling associated with the development of brain metastasis (BM) after surgical resection of NSCLC. METHODS: Rapidly frozen NSCLC surgical specimens were procured from tumor banks. RNA was extracted and analyzed by RNA-sequencing (Illumina HiSeq 2500). Clinical parameters and gene expression were examined for differentiating between patients with BM, patients with metastases to sites other than brain, and patients who did not develop metastatic disease at a clinically significant follow up. Principal component analysis and pathway enrichments studies were done. RESULTS: A total of 91 patients were included in this study, 32 of which developed BM. Stage of disease at diagnosis (P=0.004) and level of differentiation (P=0.007) were significantly different between BM and control group. We identified a set of 22 genes which correlated specifically with BM, and not with metastasis to other sites. This set achieved 93.4% accuracy (95% CI: 86.2-97.5%), 96.6% specificity and 87.5% sensitivity of correctly identifying BM patients in a leave-one-out internal validation analysis. The oxidative phosphorylation pathway was strongly correlated with BM risk. CONCLUSIONS: Expression level of a small set of genes from primary tumors was found to predict BM development, distinctly from metastasis to other organs. These genes and the correlated oxidative phosphorylation pathway require further validation as potentially clinically useful predictors of BM and possibly as novel therapeutic targets for BM prevention.

6.
Eur J Clin Microbiol Infect Dis ; 38(12): 2331-2339, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31493048

ABSTRACT

The clinical diagnosis of acute infections in the emergency department is a challenging task due to the similarity in symptom presentation between virally and bacterially infected individuals, while the use of routine laboratory tests for pathogen identification is often time-consuming and may contain contaminants. We investigated the ability of various anemia-related parameters, including hemoglobin, red cell distribution width (RDW), and iron, to differentiate between viral and bacterial infection in a retrospective study of 3883 patients admitted to the emergency department with a confirmed viral (n = 1238) or bacterial (n = 2645) infection based on either laboratory tests or microbiological cultures. The ratio between hemoglobin to RDW was found to be significant in distinguishing between virally and bacterially infected patients and outperformed other anemia measurements. Moreover, the predictive value of the ratio was high even in patients presenting with low C-reactive protein values (< 21 mg/L). We followed the dynamics of hemoglobin, RDW, and the ratio between them up to 72 h post emergency department admission, and observed a consistent discrepancy between virally and bacterially infected patients over time. Additional analysis demonstrated higher levels of ferritin and lower levels of iron in bacterially infected compared with virally infected patients. The anemia measurements were associated with length of hospital stay, where all higher levels, except for RDW, corresponded to a shorter hospitalization period. We highlighted the importance of various anemia measurements as an additional host-biomarker to discern virally from bacterially infected patients.


Subject(s)
Anemia/blood , Bacterial Infections/diagnosis , Virus Diseases/diagnosis , Anemia/microbiology , Anemia/virology , Bacterial Infections/blood , Biomarkers/blood , C-Reactive Protein/analysis , Diagnosis, Differential , Emergency Service, Hospital , Erythrocyte Indices , Ferritins/blood , Humans , Iron/blood , Length of Stay , Proportional Hazards Models , Retrospective Studies , Virus Diseases/blood
7.
Front Immunol ; 10: 1002, 2019.
Article in English | MEDLINE | ID: mdl-31130969

ABSTRACT

The host immune response against infection requires the coordinated action of many diverse cell subsets that dynamically adapt to a pathogen threat. Due to the complexity of such a response, most immunological studies have focused on a few genes, proteins, or cell types. With the development of "omic"-technologies and computational analysis methods, attempts to analyze and understand complex system dynamics are now feasible. However, the decomposition of transcriptomic data sets generated from complete organs remains a major challenge. Here, we combined Weighted Gene Coexpression Network Analysis (WGCNA) and Digital Cell Quantifier (DCQ) to analyze time-resolved mouse splenic transcriptomes in acute and chronic Lymphocytic Choriomeningitis Virus (LCMV) infections. This enabled us to generate hypotheses about complex immune functioning after a virus-induced perturbation. This strategy was validated by successfully predicting several known immune phenomena, such as effector cytotoxic T lymphocyte (CTL) expansion and exhaustion. Furthermore, we predicted and subsequently verified experimentally macrophage-CD8 T cell cooperativity and the participation of virus-specific CD8+ T cells with an early effector transcriptome profile in the host adaptation to chronic infection. Thus, the linking of gene expression changes with immune cell kinetics provides novel insights into the complex immune processes within infected tissues.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Macrophages/immunology , Transcriptome , Acute Disease , Animals , Chronic Disease , Cytokines/immunology , Gene Regulatory Networks , Male , Mice, Inbred C57BL
8.
Nat Methods ; 16(4): 327-332, 2019 04.
Article in English | MEDLINE | ID: mdl-30886410

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is a rich resource of cellular heterogeneity, opening new avenues in the study of complex tissues. We introduce Cell Population Mapping (CPM), a deconvolution algorithm in which reference scRNA-seq profiles are leveraged to infer the composition of cell types and states from bulk transcriptome data ('scBio' CRAN R-package). Analysis of individual variations in lungs of influenza-virus-infected mice reveals that the relationship between cell abundance and clinical symptoms is a cell-state-specific property that varies gradually along the continuum of cell-activation states. The gradual change is confirmed in subsequent experiments and is further explained by a mathematical model in which clinical outcomes relate to cell-state dynamics along the activation process. Our results demonstrate the power of CPM in reconstructing the continuous spectrum of cell states within heterogeneous tissues.


Subject(s)
Computational Biology , Genomics , Sequence Analysis, RNA , Single-Cell Analysis , Algorithms , Animals , Cell Separation , Female , Fibroblasts/metabolism , Flow Cytometry , Gene Expression Profiling , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Lung/virology , Markov Chains , Mice , Mice, Inbred C57BL , Orthomyxoviridae , Phagocytes/metabolism , Reference Values , Software , Transcriptome
9.
Cell Syst ; 6(6): 679-691.e4, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29886109

ABSTRACT

The influenza virus is a major cause of morbidity and mortality worldwide. Yet, both the impact of intracellular viral replication and the variation in host response across different cell types remain uncharacterized. Here we used single-cell RNA sequencing to investigate the heterogeneity in the response of lung tissue cells to in vivo influenza infection. Analysis of viral and host transcriptomes in the same single cell enabled us to resolve the cellular heterogeneity of bystander (exposed but uninfected) as compared with infected cells. We reveal that all major immune and non-immune cell types manifest substantial fractions of infected cells, albeit at low viral transcriptome loads relative to epithelial cells. We show that all cell types respond primarily with a robust generic transcriptional response, and we demonstrate novel markers specific for influenza-infected as opposed to bystander cells. These findings open new avenues for targeted therapy aimed exclusively at infected cells.


Subject(s)
Host-Pathogen Interactions/genetics , Influenza, Human/genetics , Orthomyxoviridae/genetics , Animals , Base Sequence/genetics , Cell Line , Epithelial Cells/immunology , Female , Gene Expression Profiling/methods , Host-Pathogen Interactions/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Orthomyxoviridae/metabolism , Orthomyxoviridae Infections/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics , Virus Replication
10.
Front Genet ; 7: 172, 2016.
Article in English | MEDLINE | ID: mdl-27761138

ABSTRACT

A central challenge in pharmaceutical research is to investigate genetic variation in response to drugs. The Collaborative Cross (CC) mouse reference population is a promising model for pharmacogenomic studies because of its large amount of genetic variation, genetic reproducibility, and dense recombination sites. While the CC lines are phenotypically diverse, their genetic diversity in drug disposition processes, such as detoxification reactions, is still largely uncharacterized. Here we systematically measured RNA-sequencing expression profiles from livers of 29 CC lines under baseline conditions. We then leveraged a reference collection of metabolic biotransformation pathways to map potential relations between drugs and their underlying expression quantitative trait loci (eQTLs). By applying this approach on proximal eQTLs, including eQTLs acting on the overall expression of genes and on the expression of particular transcript isoforms, we were able to construct the organization of hepatic eQTL-drug connectivity across the CC population. The analysis revealed a substantial impact of genetic variation acting on drug biotransformation, allowed mapping of potential joint genetic effects in the context of individual drugs, and demonstrated crosstalk between drug metabolism and lipid metabolism. Our findings provide a resource for investigating drug disposition in the CC strains, and offer a new paradigm for integrating biotransformation reactions to corresponding variations in DNA sequences.

11.
Bioinformatics ; 32(24): 3842-3843, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27531105

ABSTRACT

: The composition of immune-cell subsets is key to the understanding of major diseases and pathologies. Computational deconvolution methods enable researchers to investigate immune cell quantities in complex tissues based on transcriptome data. Here we present ImmQuant, a software tool allowing immunologists to upload transcription profiles of multiple tissue samples, apply deconvolution methodology to predict differences in cell-type quantities between the samples, and then inspect the inferred cell-type alterations using convenient visualization tools. ImmQuant builds on the DCQ deconvolution algorithm and allows a user-friendly utilization of this method by non-bioinformatician researchers. Specifically, it enables investigation of hundreds of immune cell subsets in mouse tissues, as well as a few dozen cell types in human samples. AVAILABILITY AND IMPLEMENTATION: ImmQuant is available for download at http://csgi.tau.ac.il/ImmQuant/ CONTACT: iritgv@post.tau.ac.ilSupplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Immune System/cytology , Software , Transcriptome , Algorithms , Animals , Humans , Mice
12.
PLoS Comput Biol ; 12(4): e1004856, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27035464

ABSTRACT

Sequence variation can affect the physiological state of the immune system. Major experimental efforts targeted at understanding the genetic control of the abundance of immune cell subpopulations. However, these studies are typically focused on a limited number of immune cell types, mainly due to the use of relatively low throughput cell-sorting technologies. Here we present an algorithm that can reveal the genetic basis of inter-individual variation in the abundance of immune cell types using only gene expression and genotyping measurements as input. Our algorithm predicts the abundance of immune cell subpopulations based on the RNA levels of informative marker genes within a complex tissue, and then provides the genetic control on these predicted immune traits as output. A key feature of the approach is the integration of predictions from various sets of marker genes and refinement of these sets to avoid spurious signals. Our evaluation of both synthetic and real biological data shows the significant benefits of the new approach. Our method, VoCAL, is implemented in the freely available R package ComICS.


Subject(s)
Algorithms , Immune System/cytology , Immune System/metabolism , Models, Genetic , Models, Immunological , Animals , Computational Biology , Gene Expression , Genetic Markers , Genotype , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Mice , Quantitative Trait Loci , Transcriptome
13.
Bioinformatics ; 31(24): 3961-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26315914

ABSTRACT

MOTIVATION: The immune system comprises a complex network of genes, cells and tissues, coordinated through signaling pathways and cell-cell communications. However, the orchestrated role of the multiple immunological components in disease is still poorly understood. Classifications based on gene-expression data have revealed immune-related signaling pathways in various diseases, but how such pathways describe the immune cellular physiology remains largely unknown. RESULTS: We identify alterations in cell quantities discriminating between disease states using ' Cell type of Disease' (CoD), a classification-based approach that relies on computational immune-cell decomposition in gene-expression datasets. CoD attains significantly higher accuracy than alternative state-of-the-art methods. Our approach is shown to recapitulate and extend previous knowledge acquired with experimental cell-quantification technologies. CONCLUSIONS: The results suggest that CoD can reveal disease-relevant cell types in an unbiased manner, potentially heralding improved diagnostics and treatment. AVAILABILITY AND IMPLEMENTATION: The software described in this article is available at http://www.csgi.tau.ac.il/CoD/.


Subject(s)
Gene Expression Profiling , Immune System/metabolism , Software , Animals , Female , Immune System/cytology , Immunity/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mice
14.
Mol Syst Biol ; 10: 720, 2014.
Article in English | MEDLINE | ID: mdl-24586061

ABSTRACT

Hundreds of immune cell types work in coordination to maintain tissue homeostasis. Upon infection, dramatic changes occur with the localization, migration, and proliferation of the immune cells to first alert the body of the danger, confine it to limit spreading, and finally extinguish the threat and bring the tissue back to homeostasis. Since current technologies can follow the dynamics of only a limited number of cell types, we have yet to grasp the full complexity of global in vivo cell dynamics in normal developmental processes and disease. Here, we devise a computational method, digital cell quantification (DCQ), which combines genome-wide gene expression data with an immune cell compendium to infer in vivo changes in the quantities of 213 immune cell subpopulations. DCQ was applied to study global immune cell dynamics in mice lungs at ten time points during 7 days of flu infection. We find dramatic changes in quantities of 70 immune cell types, including various innate, adaptive, and progenitor immune cells. We focus on the previously unreported dynamics of four immune dendritic cell subtypes and suggest a specific role for CD103(+) CD11b(-) DCs in early stages of disease and CD8(+) pDC in late stages of flu infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Influenza, Human/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CD11b Antigen/immunology , Flow Cytometry , Humans , Influenza, Human/metabolism , Influenza, Human/pathology , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Lung/immunology , Mice , Transcriptome/immunology
15.
Nat Biotechnol ; 31(4): 342-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23503680

ABSTRACT

Individual genetic variation affects gene responsiveness to stimuli, often by influencing complex molecular circuits. Here we combine genomic and intermediate-scale transcriptional profiling with computational methods to identify variants that affect the responsiveness of genes to stimuli (responsiveness quantitative trait loci or reQTLs) and to position these variants in molecular circuit diagrams. We apply this approach to study variation in transcriptional responsiveness to pathogen components in dendritic cells from recombinant inbred mouse strains. We identify reQTLs that correlate with particular stimuli and position them in known pathways. For example, in response to a virus-like stimulus, a trans-acting variant responds as an activator of the antiviral response; using RNA interference, we identify Rgs16 as the likely causal gene. Our approach charts an experimental and analytic path to decipher the mechanisms underlying genetic variation in circuits that control responses to stimuli.


Subject(s)
Dendritic Cells/virology , Gene Regulatory Networks , Genetic Variation , Quantitative Trait Loci/genetics , Transcription, Genetic , Animals , Chromosomes, Mammalian/genetics , Dendritic Cells/metabolism , Female , Gene Expression Regulation , Genetic Pleiotropy , Mice , Mice, Inbred Strains , RGS Proteins/genetics , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...