19
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Neurological Manifestations of COVID-19 Feature T Cell Exhaustion and Dedifferentiated Monocytes in Cerebrospinal Fluid

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Patients suffering from Coronavirus disease 2019 (COVID-19) can develop neurological sequelae, such as headache and neuroinflammatory or cerebrovascular disease. These conditions—termed here as Neuro-COVID—are more frequent in patients with severe COVID-19. To understand the etiology of these neurological sequelae, we utilized single-cell sequencing and examined the immune cell profiles from the cerebrospinal fluid (CSF) of Neuro-COVID patients compared with patients with non-inflammatory and autoimmune neurological diseases or with viral encephalitis. The CSF of Neuro-COVID patients exhibited an expansion of dedifferentiated monocytes and of exhausted CD4 + T cells. Neuro-COVID CSF leukocytes featured an enriched interferon signature; however, this was less pronounced than in viral encephalitis. Repertoire analysis revealed broad clonal T cell expansion and curtailed interferon response in severe compared with mild Neuro-COVID patients. Collectively, our findings document the CSF immune compartment in Neuro-COVID patients and suggest compromised antiviral responses in this setting.

          Graphical Abstract

          Highlights

          • Single-cell atlas of cerebrospinal fluid in Neuro-COVID and controls

          • Expansion of dedifferentiated monocytes and exhausted CD4 + T cells in Neuro-COVID

          • Less pronounced interferon signature in Neuro-COVID than in viral encephalitis

          • Curtailed interferon-response in severe Neuro-COVID

          Abstract

          Neurological manifestations associated with COVID-19 (Neuro-COVID) are recognized but under-studied. Using single-cell transcriptomics, Heming et al. identify expansion of dedifferentiated monocytes and exhausted CD4 + T cells in the cerebral spinal fluid of Neuro-COVID patients. This work provides a basis for improved understanding of the neurological sequelae associated with COVID-19.

          Related collections

          Most cited references85

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          A pneumonia outbreak associated with a new coronavirus of probable bat origin

          Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large number of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats 1–4 . Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans 5–7 . Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 December 2019, had caused 2,794 laboratory-confirmed infections including 80 deaths by 26 January 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence analysis of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addition, 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor—angiotensin converting enzyme II (ACE2)—as SARS-CoV.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor

            Summary The recent emergence of the novel, pathogenic SARS-coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread pose a global health emergency. Cell entry of coronaviruses depends on binding of the viral spike (S) proteins to cellular receptors and on S protein priming by host cell proteases. Unravelling which cellular factors are used by SARS-CoV-2 for entry might provide insights into viral transmission and reveal therapeutic targets. Here, we demonstrate that SARS-CoV-2 uses the SARS-CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming. A TMPRSS2 inhibitor approved for clinical use blocked entry and might constitute a treatment option. Finally, we show that the sera from convalescent SARS patients cross-neutralized SARS-2-S-driven entry. Our results reveal important commonalities between SARS-CoV-2 and SARS-CoV infection and identify a potential target for antiviral intervention.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Comprehensive Integration of Single-Cell Data

              Single-cell transcriptomics has transformed our ability to characterize cell states, but deep biological understanding requires more than a taxonomic listing of clusters. As new methods arise to measure distinct cellular modalities, a key analytical challenge is to integrate these datasets to better understand cellular identity and function. Here, we develop a strategy to "anchor" diverse datasets together, enabling us to integrate single-cell measurements not only across scRNA-seq technologies, but also across different modalities. After demonstrating improvement over existing methods for integrating scRNA-seq data, we anchor scRNA-seq experiments with scATAC-seq to explore chromatin differences in closely related interneuron subsets and project protein expression measurements onto a bone marrow atlas to characterize lymphocyte populations. Lastly, we harmonize in situ gene expression and scRNA-seq datasets, allowing transcriptome-wide imputation of spatial gene expression patterns. Our work presents a strategy for the assembly of harmonized references and transfer of information across datasets.
                Bookmark

                Author and article information

                Journal
                Immunity
                Immunity
                Immunity
                Elsevier Inc.
                1074-7613
                1097-4180
                23 December 2020
                12 January 2021
                23 December 2020
                : 54
                : 1
                : 164-175.e6
                Affiliations
                [1 ]Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
                [2 ]Department of Neurology, University Hospital Essen, Essen, Germany
                [3 ]Department of Electrical Engineering and Computer Science and Center for Computational Biology, University of California Berkeley, Berkeley, CA, USA
                [4 ]Department of Infectious Diseases, West German Centre of Infectious Diseases, University Duisburg-Essen, Germany
                [5 ]Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
                [6 ]Institute for Virology, University Hospital Essen, Germany
                [7 ]Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
                [8 ]Chan Zuckerberg Biohub, San Francisco, CA, USA
                Author notes
                []Corresponding author
                [∗∗ ]Corresponding author
                [9]

                These authors contributed equally

                [10]

                These authors contributed equally

                [11]

                Lead Contact

                Article
                S1074-7613(20)30539-2
                10.1016/j.immuni.2020.12.011
                7831653
                33382973
                bbdc1559-311f-4286-8ea2-9f136c721f6f
                © 2020 Elsevier Inc.

                Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company's public news and information website. Elsevier hereby grants permission to make all its COVID-19-related research that is available on the COVID-19 resource centre - including this research content - immediately available in PubMed Central and other publicly funded repositories, such as the WHO COVID database with rights for unrestricted research re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for free by Elsevier for as long as the COVID-19 resource centre remains active.

                History
                : 28 October 2020
                : 8 December 2020
                : 15 December 2020
                Categories
                Article

                Immunology
                transcriptomics,single-cell rna sequencing,cerebrospinal fluid,covid-19,sars-cov-2,neurological manifestation,neuro-covid,t cell exhaustion,interferon-stimulated genes

                Comments

                Comment on this article