Weekly Digests
‹ Back to December

Without CD4+ T cell help, cytotoxic effectors would not exist

December 11, 2019

Aiming to understand the mechanism behind CD4+ T cell help in regulating CD8+ T cell differentiation in the context of chronic infection and cancer, Zander and Schauder et al. utilized single-cell RNA sequencing (scRNAseq) to analyze the transcriptional profiles and differentiation pathways of CD8+ T cells during chronic viral infection. The results, recently published in Immunity, indicate that CD4+ T cells support the formation of a highly cytotoxic subset of CD8+ T cells via production of IL-21.

The researchers began by performing scRNAseq analysis on CD8+ T cells that were specific for the GP33-41 peptide of the lymphocytic choriomeningitis virus (LCMV) at 8 and 30 days post infection with the Cl13 chronic strain of LCMV. The analysis identified nine clusters, including the following three major clusters observed at Day 30:

  • The Slamf6+ subset (also found at Day 8) appeared to be a self-renewing progenitor population marked by expression of Tcf7, Id3, Icos, and Il7r, as well as an increased expression of Batf3, a transcription factor that is a critical transducer of IL-21 signaling in CD8+ T cells. This subset contained the previously identified CXCR5+TCF-1+ progenitor cells.
  • The Pdcd1+ subset appeared to comprise terminally exhausted cells with a high expression of Gzma and Gzmb, but also a high expression of inhibitory receptors such as CD244 (encoding 2B4), Cd160, Lag3, Havcr2 (encoding Tim3), and Pdcd1 (encoding PD-1). This subset was also distinguished by the expression of Cxcr6 and Cd7, as well as transcription factors Nr4a2, Maf, and Eomes.
  • The Cx3cr1+ subset appeared to have a profile similar to short-lived effectors in acute infections, characterized by a high expression of killer lectin-like receptors (Klre1, Klra9, Klrd1, and Klrg1) as well as transcription factors Tbx21 (encoding T-bet), Zeb2, Klf2, Id2, and Runx1.

An effector cluster found at Day 8 and the two unique effector-related Day 30 clusters (Pdcd1+ and Cx3cr1+; terminally exhausted and short-lived, respectively) were highly divergent from each other. However, Day 8 effectors did share expression of some genes with each of the two Day 30 clusters, indicating that by day 8, effector CD8+ T cells began to acquire characteristics of the subsets seen at day 30.

To confirm these results at the protein level and to determine tissue distribution, the researchers compared surface marker expression of the chemokine receptor CX3CR1 and Ly108 (encoded by Slamf6) among LCMV-specific CD8+ T cells in mice infected with LCMV Cl13. The Ly108+ subset (representing the Slamf6+ cluster) was mostly found in secondary lymphoid tissues (i.e. spleen, lymph nodes) and displayed a high expression of CXCR5, CD127 (encoded by Il7r), and TCF-1, as well an enhanced capacity for production of IFNγ and TNFα upon peptide stimulation. The Ly108-CX3CR1- subset (representing the Pdcd1+ cluster) was enriched in the bone marrow and liver and had an increased expression of CXCR6, Eomes, PD-1, Tim3, and 2B4. This subset was characterized by the lowest potential for pro-inflammatory cytokine production among the three analyzed subsets, consistent with exhaustion. The CX3CR1+ subset (representing the Cx3cr1+ cluster) was found predominantly in peripheral blood, lungs, and inguinal lymph nodes, and had an increased expression of KLRG1, KLRa9, T-bet, and granzyme B, as well as enhanced cytotoxicity ex vivo against peptide-pulsed target cells. Thus, the surface marker expression data were consistent with the scRNAseq gene expression results.

To determine cell differentiation trajectories, Zander and Schauder et al. applied a machine learning algorithm (Monocle 2) to the scRNAseq data. The algorithm predicted that Slamf6+ cells could terminally differentiate into either Pdcd1+ or Cx3cr1+ cells. Consistent with the algorithm predictions, adoptively transferred Ly108+CD8+ T cells gave rise to the Ly108-CX3CR1-PD-1hi and CX3CR1+ subsets in vivo. Furthermore, when all three subsets were adoptively transferred, Ly108+CD8+ T cells expanded much more than the other two subsets in the blood, spleen, and liver, and had an increased expression of Ki67. Together, these results further support the notion that the Ly108+CD8+ T cells have the self-renewing progenitor phenotype. Interestingly, while the majority of transferred CX3CR1+ cells maintained their phenotype, indicating terminal differentiation status, 40-50% of transferred Ly108-CX3CR1-PD-1hi cells acquired CX3CR1 and T-bet expression, suggesting some heterogeneity with respect to fate determination.

Given previous knowledge that CD4+ T cells play a role in sustaining CD8+ T cell responses during chronic viral infection, that sustained responses rely on CD8+ T cell-intrinsic IL-21R signaling, and that CD4+ T cells have previously been identified as a major source of IL-21, Zander and Schauder et al. explored mechanistically how CD4+ T cells play a role in CD8+ T cell differentiation during persistent viral infection. Consistent with previous observations, over 90% of IL-21-producing lymphocytes in the spleen during LCMV Cl13 infection were CD4+ T cells. Using several types of experiments, including transient depletion, adoptive cell transfer, and bone marrow chimeras, the researchers demonstrated that mechanistically, CD4+ T cell-derived IL-21 played a critical role in facilitating the formation of cytotoxic CX3CR1+CD8+ T cells, which were required for controlling viral replication during persistent viral infection in vivo. CD4+ T cell-derived IL-21 most likely promoted the differentiation of Ly108+CD8+ T cells toward CX3CR1+ cells and suppressed differentiation toward the exhausted Ly108-CX3CR1-PD-1hi phenotype. Furthermore, in an attempt to at least partially reverse T cell exhaustion, the researchers treated mice with PD-L1 blockade and found that although PD-L1 blockade reduced the viral load in mice with intact CD4+ T cells, it failed to do so in mice depleted of CD4+ T cells. Mechanistically, in the absence of CD4+ T cell help, PD-L1 blockade could not rescue the formation of the CX3CR1+CD8+ T cell subset.

Finally, the researchers examined whether tumor-infiltrating CD8+ T cells (TILs) follow a differentiation path similar to the virus-specific cells in a chronic viral infection. In B16F10 melanoma tumors, the majority of TILs were Ly108-CX3CR1-PD-1hi, consistent with a high level of exhaustion. Very few Ly108+ TILs were detected in the tumor. Adoptive transfer of tumor-specific IL-21+CD4+ T cells (with a Th17 phenotype) into B16F10 tumor-bearing mice resulted in a ~2-fold increase (compared to transfer of comparable IL-21-CD4+ T cells) in tumor-infiltrating CX3CR1+CD8+ T cells and enhanced tumor control. IL-21+CD4+ helper T cell response positively correlated with the proportion of CX3CR1+CD8+ TILs and inversely correlated with tumor burden. These effects were dependent on CD8+ T cell-intrinsic IL-21R signaling.

Overall, Zander and Schauder et al. demonstrated that in the context of a chronic viral infection or cancer, CD4+ T cell help – in the form of IL-21 production – was required for the formation of the cytotoxic CX3CR1+CD8+ T cell subset, which was responsible for the control of viral load or tumor, respectively. Furthermore, the presence of CD4+ T cell help was required for the efficacy of PD-L1 blockade. The results of this study elucidate a mechanistic pathway that could play a role in improving the efficacy of cancer-targeting immunotherapies.

by Anna Scherer

Meet the Researcher

First co-author Ryan Zander answered our questions this week.

The Cui lab


What prompted you to tackle this research question?
Recent work in the field has indicated that CD8+ T cells responding to chronic viral infection can be broadly compartmentalized into two major subsets, with a CXCR5hiTCF-1hi subset serving as a self-renewing progenitor population that can give rise to a more terminally exhausted CXCR5loTCF-1lo subset. Similar progenitor-like and exhausted CD8+ T cell subsets have also been observed during many different types of cancer. These observations led us to ask whether additional heterogeneity exists among CD8+ T cells responding to persistent infection or tumorigenesis, and whether CD4+ T cell help regulates this multifaceted process of CD8+ T cell differentiation.

What was the most surprising finding of this study for you?

I think that the most surprising finding of this study was the identification of a transcriptionally distinct CD8+ T cell subset that displays potent cytolytic activity, and whose development is wholly dependent on CD4+ T cell help. Although CD4+ T cell help has long been known to be important for sustaining CD8+ T cell responses, the underlying mechanisms by which this occurs have remained unclear. Our discovery that a lack of CD4+ T cell help largely precludes the differentiation of a functional cytotoxic CD8+ T cell subset is really quite striking and has important implications for immunotherapy.

What was the coolest thing you’ve learned (about) recently outside of work?

A recent Snapple fact that I found to be quite entertaining is that flamingos are actually born with grey feathers that then gradually turn pink due to a dye that they obtain from their diet of shrimp.

References:

Zander R., Schauder D., Xin G., Nguyen C., Wu X., Zajac A., Cui W. CD4(+) T Cell Help Is Required for the Formation of a Cytolytic CD8(+) T Cell Subset that Protects against Chronic Infection and Cancer. Immunity. 2019 Nov 27.

In the Spotlight...

Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation

Wu and Seung et al. developed CD38/CD3×CD28 trispecific IgG4s mutated to negate FcR binding and non-specific cytokine release. Crystal structures showed solvent-exposed CDRs and modeling showed trispecific molecules binding opposite each other to CD28 dimer. In vitro, each variable fragment contributed to human TCM, TH1, and antigen-specific T cell proliferation. CD28 on CD38+ myeloma cells enhanced targeting and contributed to trispecific antibody-mediated human T cell lysis of myeloma cells in vitro and myeloma growth inhibition in a humanized mouse model. Immune effects were observed in non-human primates at tolerated doses.

Contributed by Paula Hochman

Proliferating Transitory T Cells with an Effector-like Transcriptional Signature Emerge from PD-1+ Stem- like CD8+ T Cells during Chronic Infection

Following an RNAseq-based targeted search for novel CD8+ T cell surface markers of exhaustion in the well-studied LCMV system, Hudson et al. found that CD101 expression could be used to distinguish two trajectory-related populations following stimulation of antigen-specific PD-1+Tcf-1+ stem cells. A CD101-Tim3+CX3CR1+ population arose first, characterized by high proliferative capacity, functional capability, and ability to control viral load, followed in 2-4 weeks by CD101+Tim3+CX3CR1- terminally exhausted cells with low proliferative and functional capacity. PD-1 blockade significantly enhanced expansion of the effector CD101- population.

PD-1 Signaling Promotes Control of Chronic Viral Infection by Restricting Type-I-Interferon-Mediated Tissue Damage

Raju et al. carried out a time-dependent evaluation of PD-1 blockade on tissue damage, CD8+ T cell function, and viral control following chronic LCMV infection. Acute PD-1 blockade (days 0–8) with anti-PD-L1 was uniformly fatal; subacute blockade (days 8–22) resulted in survival, persistent viremia, lymphoid organ pathology, and a transient increase in CD8+ LCMV-specific T cells. Anti-IFNAR with acute anti-PD-L1 prevented death and increased the frequency of LCMV-specific T cells. Anti-IFNAR followed by subacute anti-PD-L1 restored CD8+ T cells and accelerated viral clearance, suggesting a protective role for PD-1 by antagonizing Type 1 IFN immunopathology.

Contributed by Katherine Turner

TCR sequencing paired with massively parallel 3' RNA-seq reveals clonotypic T cell signatures

To couple TCR CDR3 identification with gene expression phenotyping in individual cells, Tu and Gierahn et al. developed a PCR strategy to amplify 3’-barcoded individual whole cell transcriptomes, followed by TCR gene enrichment and amplification prior to targeted Illumina sequencing of the 3’-barcode and the CDR3. In ~20% of the cells, both CDR3s could be identified and matched to the barcoded mRNA phenotype. Application to a mouse model antigen immunization revealed clonotypic effects on CD8+ T cell phenotype; application to human CD4+ T cells from patients with peanut allergy revealed expanded clonotypes with a TH2 phenotype.

Transposable element expression in tumors is associated with immune infiltration and increased antigenicity

To analyze transposable element (TE) expression in the genome, Kong et al. produced a computational method REdiscoverTE. In TCGA and Cancer Genome Project data sets, various TE subfamilies, particularly from evolutionarily young TEs, were overexpressed in tumor tissue based on the cancer type. TE expression correlated with proximal DNA demethylation and expression of immune and DNA damage response genes. In glioblastoma cell lines, reversing DNA methylation with decitabine increased expression of TEs and TE-derived peptides detected in the proteome and MHC peptidome for potential recognition by antitumor T cells.

Contributed by Alex Najibi

Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors

To advance utilization of allogeneic T cells for CAR therapy, Webber and Lonetree et al. showed that the CRISPR-CAS9 base editing technology could overcome the undesirable effects of double-strand breaks initiated with CAS9 nuclease, and focused on altering critical splice sites to eliminate gene expression. Maximal efficiency was observed with codon-optimized mRNA encoding the editing machinery. Simultaneous multiplex knockdout of the TRAC, β2M, and PDCD1 (encoding PD-1) loci in primary human T cells approached 90% efficiency; the cells retained polyfunctionality and cytotoxicity, including toward a PD-L1-overexpressing target.

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.