skip to main content


Title: Interrogating Adaptive Immunity Using LCMV
Abstract

In this invited article, we explain technical aspects of the lymphocytic choriomeningitis virus (LCMV) system, providing an update of a prior contribution by Matthias von Herrath and J. Lindsay Whitton. We provide an explanation of the LCMV infection models, highlighting the importance of selecting an appropriate route and viral strain. We also describe how to quantify virus‐specific immune responses, followed by an explanation of useful transgenic systems. Specifically, our article will focus on the following protocols. © 2020 Wiley Periodicals LLC.

Basic Protocol 1: LCMV infection routes in mice

Support Protocol 1: Preparation of LCMV stocks

ASSAYS TO MEASURE LCMV TITERS

Support Protocol 2: Plaque assay

Support Protocol 3: Immunofluorescence focus assay (IFA) to measure LCMV titer

MEASUREMENT OF T CELL AND B CELL RESPONSES TO LCMV INFECTION

Basic Protocol 2: Triple tetramer staining for detection of LCMV‐specific CD8 T cells

Basic Protocol 3: Intracellular cytokine staining (ICS) for detection of LCMV‐specific T cells

Basic Protocol 4: Enumeration of direct ex vivo LCMV‐specific antibody‐secreting cells (ASC)

Basic Protocol 5: Limiting dilution assay (LDA) for detection of LCMV‐specific memory B cells

Basic Protocol 6: ELISA for quantification of LCMV‐specific IgG antibody

Support Protocol 4: Preparation of splenic lymphocytes

Support Protocol 5: Making BHK21‐LCMV lysate

Basic Protocol 7: Challenge models

TRANSGENIC MODELS

Basic Protocol 8: Transfer of P14 cells to interrogate the role of IFN‐I on CD8 T cell responses

Basic Protocol 9: Comparing the expansion of naïve versus memory CD4 T cells following chronic viral challenge

 
more » « less
NSF-PAR ID:
10168877
Author(s) / Creator(s):
 ;  ;  ;  
Publisher / Repository:
Wiley Blackwell (John Wiley & Sons)
Date Published:
Journal Name:
Current Protocols in Immunology
Volume:
130
Issue:
1
ISSN:
1934-3671
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. Abstract

    Antigen‐specific memory B cell (MBC) populations mediate the rapid, strong, and high‐affinity secondary antibody responses that play a key role in combating infection and generating protective responses to vaccination. Recently, cell staining with fluorochrome‐labeled antigens together with sequencing methods such as Drop‐seq and CITE‐seq have provided information on the specificity, phenotype, and transcriptome of single MBCs. However, characterization of MBCs at the level of antigen‐reactive populations remains an important tool for assessing an individual's B cell immunity and responses to antigen exposure. This is readily performed using a long‐established method based on in vitro polyclonal stimulation of MBCs to induce division and differentiation into antibody‐secreting cells (ASCs). Post‐stimulation antigen‐specific measurement of the MBC‐derived ASCs (or the secreted antibodies) indicates the size of precursor MBC populations. Additional information about the character of antigen‐reactive MBC populations is provided by analysis of MBC‐derived antibodies of particular specificities for binding avidity and functionality. This article outlines a simple and reliable strategy for efficient in vitro MBC stimulation and use of the ELISpot assay as a post‐stimulation readout to determine the size of antigen‐specific MBC populations. Other applications of the in vitro stimulation technique for MBC analysis are discussed. The following protocols are included. © 2020 Wiley Periodicals LLC

    Basic Protocol 1: Polyclonal stimulation of memory B cells using unfractionated PBMCs

    Alternate Protocol: Stimulation of small PBMC numbers using 96‐well plates with U‐bottom wells

    Basic Protocol 2: ELISpot assay for enumeration of memory B cell−derived antibody‐secreting cells

     
    more » « less
  2. Abstract

    Class II major histocompatibility complex peptide (MHC‐IIp) multimers are precisely engineered reagents used to detect T cells specific for antigens from pathogens, tumors, and self‐proteins. While the related Class I MHC/peptide (MHC‐Ip) multimers are usually produced from subunits expressed inE. coli, most Class II MHC alleles cannot be produced in bacteria, and this has contributed to the perception that MHC‐IIp reagents are harder to produce. Herein, we present a robust constitutive expression system for soluble biotinylated MHC‐IIp proteins that uses stable lentiviral vector−transduced derivatives of HEK‐293T cells. The expression design includes allele‐specific peptide ligands tethered to the amino‐terminus of the MHC‐II β chain via a protease‐cleavable linker. Following cleavage of the linker, HLA‐DM is used to catalyze efficient peptide exchange, enabling high‐throughput production of many distinct MHC‐IIp complexes from a single production cell line. Peptide exchange is monitored using either of two label‐free methods, native isoelectric focusing gel electrophoresis or matrix‐assisted laser desorption/ionization time‐of‐flight (MALDI‐TOF) mass spectrometry of eluted peptides. Together, these methods produce MHC‐IIp complexes that are highly homogeneous and that form the basis for excellent MHC‐IIp multimer reagents. © 2021 Wiley Periodicals LLC.

    This article was corrected on 19 July 2022. See the end of the full text for details.

    Basic Protocol 1: Lentivirus production and expression line creation

    Support Protocol 1: Six‐well assay for estimation of production cell line yield

    Support Protocol 2: Universal ELISA for quantifying proteins with fused leucine zippers and His‐tags

    Basic Protocol 2: Cultures for production of Class II MHC proteins

    Basic Protocol 3: Purification of Class II MHC proteins by anti‐leucine zipper affinity chromatography

    Alternate Protocol 1: IMAC purification of His‐tagged Class II MHC

    Support Protocol 3: Protein concentration measurements and adjustments

    Support Protocol 4: Polishing purification by anion‐exchange chromatography

    Support Protocol 5: Estimating biotinylation percentage by streptavidin precipitation

    Basic Protocol 4: Peptide exchange

    Basic Protocol 5: Analysis of peptide exchange by matrix‐assisted laser desorption/ionization (MALDI) mass spectrometry

    Alternate Protocol 2: Native isoelectric focusing to validate MHC‐II peptide loading

    Basic Protocol 6: Multimerization

    Basic Protocol 7: Staining cells with Class II MHC tetramers

     
    more » « less
  3. Abstract

    Staphylococcus aureusis a Gram‐positive bacterium that colonizes almost every organ in humans and mice and is a leading cause of diseases worldwide.S. aureusinfections can be challenging to treat due to widespread antibiotic resistance and their ability to cause tissue damage. The primary modes of transmission ofS. aureusare via direct contact with a colonized or infected individual or invasive spread from a colonization niche in the same individual.S. aureuscan cause a myriad of diseases, including skin and soft tissue infections (SSTIs), osteomyelitis, pneumonia, endocarditis, and sepsis.S. aureusinfection is characterized by the formation of purulent lesions known as abscesses, which are rich in live and dead neutrophils, macrophages, and surrounded by a capsule containing fibrin and collagen. Different strains ofS. aureusproduce varying amounts of toxins that evade and/or elicit immune responses. Therefore, animal models ofS. aureusinfection provide a unique opportunity to understand the dynamics of organ‐specific immune responses and modifications in the pathogen that could favor the establishment of the pathogen. With advances in in vivo imaging of fluorescent transgenic mice, combined with fluorescent/bioluminescent bacteria, we can use mouse models to better understand the immune response to these types of infections. By understanding the host and bacterial dynamics within various organ systems, we can develop therapeutics to eliminate these pathogens. This module describes in vivo mouse models of both local and systemicS. aureusinfection. © 2021 Wiley Periodicals LLC.

    This article was corrected on 20 July 2022. See the end of the full text for details.

    Basic Protocol 1: Murine model ofStaphylococcus aureussubcutaneous infection

    Alternate Protocol: Murine tape stripping skin infection model

    Basic Protocol 2: Sample collection to determine skin structure, production of inflammatory mediators, and bacterial load

    Basic Protocol 3: Murine model of post‐traumaticStaphylococcus aureusosteomyelitis

    Basic Protocol 4: Intravenous infection of the retro‐orbital sinus

    Support Protocol: Preparation of the bacterial inoculum

     
    more » « less
  4. Abstract

    New genetic engineering techniques have advanced the field of plant molecular biology, andAgrobacterium‐mediated transformation has enabled the discovery of numerous molecular and genetic functions. It has been widely used in many plants, including the economically important crop, soybean. Large‐scale genetic analyses are needed to comprehend the molecular mechanisms that underlie the agronomic traits of soybean, and the generation of stable transgenic plants involves a lengthy and laborious process.Agrobacterium rhizogenes‐mediated hairy root transformation is a quick and efficient method for investigations of root‐specific processes and interactions. Generation of composite plants with transgenic roots and wild‐type shoots allows for the study of the genetic mechanisms involved in root biology, such as theBradyrhizobium‐soybean interaction. Here, we provide an updated protocol for generating hairy soybean roots in as little as 18 days in a cost‐ and space‐effective manner and demonstrate possible uses of composite plants with soybean nodulation assays and gene expression analysis as examples. © 2021 Wiley Periodicals LLC.

    Basic Protocol 1: Soybean hairy root transformation

    Basic Protocol 2: Soybean nodulation assay

    Alternate Protocol: Soybean nodulation assay in germination pouches

    Support Protocol:Bradyrhizobium japonicumculture preparation for inoculation

    Basic Protocol 3: Histochemical GUS staining for promoter analysis

     
    more » « less
  5. Abstract

    Cross‐presentation was first observed serendipitously in the 1970s. The importance of it was quickly realized and subsequently attracted great attention from immunologists. Since then, our knowledge of the ability of certain antigen presenting cells to internalize, process, and load exogenous antigens onto MHC‐I molecules to cross‐prime CD8+T cells has increased significantly. Dendritic cells (DCs) are exceptional cross‐presenters, thus making them a great tool to study cross‐presentation but the relative rarity of DCs in circulation and in tissues makes it challenging to isolate sufficient numbers of cells to study this process in vitro. In this paper, we describe in detail two methods to culture DCs from bone‐marrow progenitors and a method to expand the numbers of DCs present in vivo as a source of endogenous bona‐fide cross‐presenting DCs. We also describe methods to assess cross‐presentation by DCs using the activation of primary CD8+T cells as a readout. © 2020 Wiley Periodicals LLC.

    Basic Protocol 1: Isolation of bone marrow progenitor cells

    Basic Protocol 2: In vitro differentiation of dendritic cells with GM‐CSF

    Support Protocol 1: Preparation of conditioned medium from GM‐CSF producing J558L cells

    Basic Protocol 3: In vitro differentiation of dendritic cells with Flt3L

    Support Protocol 2: Preparation of Flt3L containing medium from B16‐Flt3L cells

    Basic Protocol 4: Expansion of cDC1s in vivo for use in ex vivo experiments

    Basic Protocol 5: Characterizing resting and activated dendritic cells

    Basic Protocol 6: Dendritic cell stimulation, antigenic cargo, and fixation

    Support Protocol 3: Preparation of model antigen coated microbeads

    Support Protocol 4: Preparation of apoptotic cells

    Support Protocol 5: Preparation of recombinant bacteria

    Basic Protocol 7: Immunocytochemistry immunofluorescence (ICC/IF)

    Support Protocol 6: Preparation of Alcian blue‐coated coverslips

    Basic Protocol 8: CD8+T cell activation to assess cross‐presentation

    Support Protocol 7: Isolation and labeling of CD8+T cells with CFSE

     
    more » « less