Intestinal allograft tolerance in large animals

  • Funded by National Institutes of Health (NIH)
  • Total publications:0 publications

Grant number: unknown

Grant search

Key facts

  • Disease

    COVID-19
  • Start & end year

    2020
    2022
  • Known Financial Commitments (USD)

    $283,116
  • Funder

    National Institutes of Health (NIH)
  • Principal Investigator

    Pending
  • Research Location

    United States of America
  • Lead Research Institution

    COLUMBIA UNIVERSITY HEALTH SCIENCES
  • Research Priority Alignment

    N/A
  • Research Category

    Pathogen: natural history, transmission and diagnostics

  • Research Subcategory

    Immunity

  • Special Interest Tags

    N/A

  • Study Subject

    Non-Clinical

  • Clinical Trial Details

    N/A

  • Broad Policy Alignment

    Pending

  • Age Group

    Not Applicable

  • Vulnerable Population

    Not applicable

  • Occupations of Interest

    Not applicable

Abstract

Project Summary: There is growing evidence that an overexuberant innate and adaptive immune responsemay contribute to life-threatening pulmonary pathology in COVID-19 disease. On the other hand, inadequateviral control may allow severe disease to develop. Human immune system (HIS) mouse models have enormousand unique potential to model human COVID-19. Unlike other animal models, HIS mice could be used tounderstand the role of the innate and adaptive human immune systems in both controlling and driving SARS-CoV2-mediated disease and hence be used to optimize therapeutic approaches. The goal of our proposal is tooptimize HIS mouse models for these purposes. Specifically, we propose to: 1) Optimize our HIS mousemodels for the study of COVID-19. Existing mouse models are limited by the lack of human ACE2, the SARS-CoV2 receptor, in the respiratory tract. We will implant iPS cell-derived human lung bud organoids generatedfrom cord blood HSC donor cells into HLA-A2 Tg NSG mice receiving HLA-A2+ cord blood HSCs. In a secondapproach to humanizing mice for COVID-19 mouse studies, we will use CRISPR/Cas9 to replace the murineACE2 gene with hACE2, allowing physiologic expression of hACE2 in NSG mice. Human HSC recipients will betreated with mouse TSLP in an AAV vector to enhance murine thymic and lymph node structure and therebyimprove human T cell development and improve peripheral vaccination responses. An alternative approach toenhancing thymus function will involve grafting of multiple pieces of thymocyte-depleted neonatal human thymustissue in multiple sites to compensate for the lack of growth potential (compared to fetal thymus) of neonatalthymus tissue. Immune reconstitution, T cell reconstitution and lymphoid structure will be followed and humoraland cellular responses to live attenuated SARS-CoV2 virus vaccination will be measured; 2) Use optimized HISmouse models to determine the kinetics of disease pathogenesis and the role of human immunecomponents in controlling infection and mediating pathologic host responses. We will first employ HISmice constructed with human cord blood HSCs and autologous iPSC-derived lung bud implants and later utilizethe above HLA-A2 hACE2 Tg model. Baseline infection with SARS-CoV2 will be assessed in non-reconstitutedanimals and compared to HIS mice. In HIS mice, we will deplete various human immune components (T cells,B cells or macrophages) to determine their impact on the course of infection and pathology associated withSARS-CoV2. In hACE2 Tg mice we will investigate the kinetics of infection of various components of therespiratory tract in combination with analysis of the human immune cell infiltrates in each locale over time. Withthese models established, we will be positioned to test therapeutic approaches during different phasesof SARS-CoV2 infection in future studies. Collectively, our models will provide critical information on the roleof human immune components in driving and protecting from COVID-19-associated pathology, allowingaccelerated optimization of immunomodulatory therapies.