Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-514070

RESUMO

The molecular underpinnings of organ dysfunction in acute COVID-19 and its potential long-term sequelae are under intense investigation. To shed light on these in the context of liver function, we performed single-nucleus RNA-seq and spatial transcriptomic profiling of livers from 17 COVID-19 decedents. We identified hepatocytes positive for SARS-CoV-2 RNA with an expression phenotype resembling infected lung epithelial cells. Integrated analysis and comparisons with healthy controls revealed extensive changes in the cellular composition and expression states in COVID-19 liver, reflecting hepatocellular injury, ductular reaction, pathologic vascular expansion, and fibrogenesis. We also observed Kupffer cell proliferation and erythrocyte progenitors for the first time in a human liver single-cell atlas, resembling similar responses in liver injury in mice and in sepsis, respectively. Despite the absence of a clinical acute liver injury phenotype, endothelial cell composition was dramatically impacted in COVID-19, concomitantly with extensive alterations and profibrogenic activation of reactive cholangiocytes and mesenchymal cells. Our atlas provides novel insights into liver physiology and pathology in COVID-19 and forms a foundational resource for its investigation and understanding.

2.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-480515

RESUMO

COVID-19 survivors develop post-acute sequelae of SARS-CoV-2 (PASC), but the mechanistic basis of PASC-associated lung abnormalities suffers from a lack of longitudinal samples. Mouse-adapted SARS-CoV-2 MA10 produces an acute respiratory distress syndrome (ARDS) in mice similar to humans. To investigate PASC pathogenesis, studies of MA10-infected mice were extended from acute disease through clinical recovery. At 15-120 days post-virus clearance, histologic evaluation identified subpleural lesions containing collagen, proliferative fibroblasts, and chronic inflammation with tertiary lymphoid structures. Longitudinal spatial transcriptional profiling identified global reparative and fibrotic pathways dysregulated in diseased regions, similar to human COVID-19. Populations of alveolar intermediate cells, coupled with focal upregulation of pro-fibrotic markers, were identified in persistently diseased regions. Early intervention with antiviral EIDD-2801 reduced chronic disease, and early anti-fibrotic agent (nintedanib) intervention modified early disease severity. This murine model provides opportunities to identify pathways associated with persistent SARS-CoV-2 pulmonary disease and test countermeasures to ameliorate PASC.

3.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-434433

RESUMO

The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) virus has infected over 115 million people and caused over 2.5 million deaths worldwide. Yet, the molecular mechanisms underlying the clinical manifestations of COVID-19, as well as what distinguishes them from common seasonal influenza virus and other lung injury states such as Acute Respiratory Distress Syndrome (ARDS), remains poorly understood. To address these challenges, we combined transcriptional profiling of 646 clinical nasopharyngeal swabs and 39 patient autopsy tissues, matched with spatial protein and expression profiling (GeoMx) across 357 tissue sections. These results define both body-wide and tissue-specific (heart, liver, lung, kidney, and lymph nodes) damage wrought by the SARS-CoV-2 infection, evident as a function of varying viral load (high vs. low) during the course of infection and specific, transcriptional dysregulation in splicing isoforms, T cell receptor expression, and cellular expression states. In particular, cardiac and lung tissues revealed the largest degree of splicing isoform switching and cell expression state loss. Overall, these findings reveal a systemic disruption of cellular and transcriptional pathways from COVID-19 across all tissues, which can inform subsequent studies to combat the mortality of COVID-19, as well to better understand the molecular dynamics of lethal SARS-CoV-2 infection and other viruses.

4.
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.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-430130

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...