Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
medrxiv; 2024.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2024.02.02.24302068

ABSTRACT

Asthma is a complex disease caused by genetic and environmental factors. Epidemiological studies have shown that in children, wheezing during rhinovirus infection (a cause of the common cold) is associated with asthma development during childhood. This has led scientists to hypothesize there could be a causal relationship between rhinovirus infection and asthma or that RV-induced wheezing identifies individuals at increased risk for asthma development. However, not all children who wheeze when they have a cold develop asthma. Genome-wide association studies (GWAS) have identified hundreds of genetic variants contributing to asthma susceptibility, with the vast majority of likely causal variants being non-coding. Integrative analyses with transcriptomic and epigenomic datasets have indicated that T cells drive asthma risk, which has been supported by mouse studies. However, the datasets ascertained in these integrative analyses lack airway epithelial cells. Furthermore, large-scale transcriptomic T cell studies have not identified the regulatory effects of most non-coding risk variants in asthma GWAS, indicating there could be additional cell types harboring these 'missing regulatory effects'. Given that airway epithelial cells are the first line of defense against rhinovirus, we hypothesized they could be mediators of genetic susceptibility to asthma. Here we integrate GWAS data with transcriptomic datasets of airway epithelial cells subject to stimuli that could induce activation states relevant to asthma. We demonstrate that epithelial cultures infected with rhinovirus significantly upregulate childhood-onset asthma-associated genes. We show that this upregulation occurs specifically in non-ciliated epithelial cells. This enrichment for genes in asthma risk loci, or 'asthma heritability enrichment' is also significant for epithelial genes upregulated with influenza infection, but not with SARS-CoV-2 infection or cytokine activation. Additionally, cells from patients with asthma showed a stronger heritability enrichment compared to cells from healthy individuals. Overall, our results suggest that rhinovirus infection is an environmental factor that interacts with genetic risk factors through non-ciliated airway epithelial cells to drive childhood-onset asthma.


Subject(s)
Genomic Instability , Infections , Asthma , COVID-19 , Influenza, Human
2.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.03.19.436212

ABSTRACT

ABSTRACT Genome-wide association studies (GWAS) provide a powerful means to identify loci and genes contributing to disease, but in many cases the related cell types/states through which genes confer disease risk remain unknown. Deciphering such relationships is important for identifying pathogenic processes and developing therapeutics. Here, we introduce sc-linker, a framework for integrating single-cell RNA-seq (scRNA-seq), epigenomic maps and GWAS summary statistics to infer the underlying cell types and processes by which genetic variants influence disease. We analyzed 1.6 million scRNA-seq profiles from 209 individuals spanning 11 tissue types and 6 disease conditions, and constructed gene programs capturing cell types, disease progression, and cellular processes both within and across cell types. We evaluated these gene programs for disease enrichment by transforming them to SNP annotations with tissue-specific epigenomic maps and computing enrichment scores across 60 diseases and complex traits (average N= 297K). Cell type, disease progression, and cellular process programs captured distinct heritability signals even within the same cell type, as we show in multiple complex diseases that affect the brain (Alzheimer’s disease, multiple sclerosis), colon (ulcerative colitis) and lung (asthma, idiopathic pulmonary fibrosis, severe COVID-19). The inferred disease enrichments recapitulated known biology and highlighted novel cell-disease relationships, including GABAergic neurons in major depressive disorder (MDD), a disease progression M cell program in ulcerative colitis, and a disease-specific complement cascade process in multiple sclerosis. In autoimmune disease, both healthy and disease progression immune cell type programs were associated, whereas for epithelial cells, disease progression programs were most prominent, perhaps suggesting a role in disease progression over initiation. Our framework provides a powerful approach for identifying the cell types and cellular processes by which genetic variants influence disease.


Subject(s)
Autoimmune Diseases , Alzheimer Disease , Colitis, Ulcerative , Depressive Disorder , Multiple Sclerosis , Idiopathic Pulmonary Fibrosis , COVID-19 , Depressive Disorder, Major
3.
Toni M. Delorey; Carly G. K. Ziegler; Graham Heimberg; Rachelly Normand; Yiming Yang; Asa Segerstolpe; Domenic Abbondanza; Stephen J. Fleming; Ayshwarya Subramanian; Daniel T. Montoro; Karthik A. Jagadeesh; Kushal Dey; Pritha Sen; Michal Slyper; Yered Pita-Juarez; Devan Phillips; Zohar Bloom-Ackermann; Nick Barkas; Andrea Ganna; James Gomez; Erica Normandin; Pourya Naderi; Yury V. Popov; Siddharth S. Raju; Sebastian Niezen; Linus T.-Y. Tsai; Katherine J. Siddle; Malika Sud; Victoria M. Tran; Shamsudheen Karuthedath Vellarikkal; Liat Amir-Zilberstein; Joseph M Beechem; Olga R. Brook; Jonathan Chen; Prajan Divakar; Phylicia Dorceus; Jesse M Engreitz; Adam Essene; Donna M. Fitzgerald; Robin Fropf; Steven Gazal; Joshua Gould; Tyler Harvey; Jonathan Hecht; Tyler Hether; Judit Jane-Valbuena; Michael Leney-Greene; Hui Ma; Cristin McCabe; Daniel E. McLoughlin; Eric M. Miller; Christoph Muus; Mari Niemi; Robert Padera; Liuliu Pan; Deepti Pant; Jenna Pfiffner-Borges; Christopher J. Pinto; Jason Reeves; Marty Ross; Melissa Rudy; Erroll H. Rueckert; Michelle Siciliano; Alexander Sturm; Ellen Todres; Avinash Waghray; Sarah Warren; Shuting Zhang; Dan Zollinger; Lisa Cosimi; Rajat M Gupta; Nir Hacohen; Winston Hide; Alkes L. Price; Jayaraj Rajagopal; Purushothama Rao Tata; Stefan Riedel; Gyongyi Szabo; Timothy L. Tickle; Deborah Hung; Pardis C. Sabeti; Richard Novak; Robert Rogers; Donald E. Ingber; Z Gordon Jiang; Dejan Juric; Mehrtash Babadi; Samouil L. Farhi; James R. Stone; Ioannis S. Vlachos; Isaac H. Solomon; Orr Ashenberg; Caroline B.M. Porter; Bo Li; Alex K. Shalek; Alexandra-Chloe Villani; Orit Rozenblatt-Rosen; Aviv Regev.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.02.25.430130

ABSTRACT

The SARS-CoV-2 pandemic has caused over 1 million deaths globally, mostly due to acute lung injury and acute respiratory distress syndrome, or direct complications resulting in multiple-organ failures. Little is known about the host tissue immune and cellular responses associated with COVID-19 infection, symptoms, and lethality. To address this, we collected tissues from 11 organs during the clinical autopsy of 17 individuals who succumbed to COVID-19, resulting in a tissue bank of approximately 420 specimens. We generated comprehensive cellular maps capturing COVID-19 biology related to patients demise through single-cell and single-nucleus RNA-Seq of lung, kidney, liver and heart tissues, and further contextualized our findings through spatial RNA profiling of distinct lung regions. We developed a computational framework that incorporates removal of ambient RNA and automated cell type annotation to facilitate comparison with other healthy and diseased tissue atlases. In the lung, we uncovered significantly altered transcriptional programs within the epithelial, immune, and stromal compartments and cell intrinsic changes in multiple cell types relative to lung tissue from healthy controls. We observed evidence of: alveolar type 2 (AT2) differentiation replacing depleted alveolar type 1 (AT1) lung epithelial cells, as previously seen in fibrosis; a concomitant increase in myofibroblasts reflective of defective tissue repair; and, putative TP63+ intrapulmonary basal-like progenitor (IPBLP) cells, similar to cells identified in H1N1 influenza, that may serve as an emergency cellular reserve for severely damaged alveoli. Together, these findings suggest the activation and failure of multiple avenues for regeneration of the epithelium in these terminal lungs. SARS-CoV-2 RNA reads were enriched in lung mononuclear phagocytic cells and endothelial cells, and these cells expressed distinct host response transcriptional programs. We corroborated the compositional and transcriptional changes in lung tissue through spatial analysis of RNA profiles in situ and distinguished unique tissue host responses between regions with and without viral RNA, and in COVID-19 donor tissues relative to healthy lung. Finally, we analyzed genetic regions implicated in COVID-19 GWAS with transcriptomic data to implicate specific cell types and genes associated with disease severity. Overall, our COVID-19 cell atlas is a foundational dataset to better understand the biological impact of SARS-CoV-2 infection across the human body and empowers the identification of new therapeutic interventions and prevention strategies.


Subject(s)
Fibrosis , Adenocarcinoma, Bronchiolo-Alveolar , Respiratory Distress Syndrome , Acute Lung Injury , COVID-19
SELECTION OF CITATIONS
SEARCH DETAIL