Weekly Digests
‹ Back to May

MDSCs play metabolic freeze tag with T cells

May 13, 2020

Suppressive myeloid cells, namely myeloid-derived suppressor cells (MDSCs), are known to appear in situations of chronic inflammation and cancer, where they suppress cytotoxic T cell responses. However, without an existing definitive marker for this immune subset, MDSCs can be difficult to study and target. In research recently published in Nature Immunology, Baumann et al. not only present evidence of a metabolic marker for MDSCs, but also show how that marker plays a role in the suppression of effector T cell functions.

To create a model of MDSCs to study, Baumann et al. used culturing with a human stromal fibroblast cell line to convert human peripheral blood monocytes into MDSCs that were phenotypically similar to those found in cancer. Transcriptome analysis showed 200 differentially expressed genes between the monocytes and MDSCs, and while it did not reveal a singular marker that was sufficient to differentiate the two cell populations, it did reveal downregulation of genes encoding glycolysis-related enzymes in MDSCs. In vitro, model MDSCs showed reduced glucose uptake, reduced surface expression of Glut-1 (the transporter that mediates glucose uptake in immune cells), lower hexokinase activity, an inability to utilize glucose for glycolysis, reduced mitochondrial membrane potential, and lower baseline mitochondrial respiration. Reduced glucose uptake and hexokinase activity were confirmed in human tumor-derived MDSCs. Together this showed that cellular metabolism is strongly reduced in MDSCs.

Looking into how in vitro-generated MDSCs interact with T cells, the researchers cocultured the two cell types and found that following interactions with MDSCs, T cells had reduced phosphorylation of key protein kinases downstream of TCR activation, suggesting suppression of TCR signaling. TCR and CD28 signaling typically increase glucose uptake and glycolysis within T cells, however, when T cells were stimulated with anti-CD3 and anti-CD28 in the presence of MDSCs, they failed to increase Glut-1 surface expression, glucose uptake, hexokinase activity, glycolysis, or mitochondrial respiration. As a result, ATP concentrations were reduced, and the T cells were unable to proliferate, express cytokines (including IFNγ and TNF), or release granzyme B. Tumor-derived MDSCs, but not monocytes, had the same suppressive effect, essentially rendering T cells unable to respond to activation.

Next, Baumann et al. observed that MDSCs were not able to suppress T cell activation when the two cell types were separated by a transwell, suggesting that the suppressive activity depends on direct cellular contact. Further investigating this interaction, they used dyes to label various cellular components of MDSCs and found that cytosolic constituents (but not entire organelles) from MDSCs transferred into nearby CD8+ T cells, as well as CD4+ T cells and NK cells, though no transfer of surface molecules was observed. Similar results were observed in tumor-derived MDSCs and an in vivo tumor model.

Investigating how the interaction between MDSCs and T cells halts T cell activation, Baumann et al. found that dimethylbiguanide (DMBG) reversed the MDSC-mediated suppression of T cell proliferation and reinstalled T cell signaling. Based on knowledge that guanidine neutralizes dicarbonyls, the researchers then searched for this class of metabolites within MDSCs and identified an accumulation of methyl­glyoxal in both model and patient-derived MDSCs. The accumulation of methylglyoxal was found to be due to semicarbazide-sensitive amine oxidase (SSAO) acting on aminoacetone. Importantly, the accumulation of methyl­glyoxal was highly specific to MDSCs and was not found in other human immune cell populations, suggesting that it could be used as a clear molecular identifier for MDSCs. When model MDSCs were incubated with DMBG, the accumulation of methyl­glyoxal was lost, the capacity to take up glucose was regained, and glycolysis and mitochondrial respiration were restored, suggesting that methylglyoxal also plays an important functional role in MDSC metabolism.

Using a methylglyoxal-sensitive fluorescent reporter to investigate whether methylglyoxal was involved in T cell suppression by MDSCs, the researchers found that methylglyoxal could be detected in CD8+ T cells within ten minutes of being in contact with MDSCs. When MDSCs were pre-treated with DMBG, however, methylglyoxal was not detected in T cells, and T cells remained capable of taking up glucose. Further, pretreating either MDSCs or CD8+ T cells with DMBG prevented the suppression of activation-induced cytokine production and granzyme B release in CD8+ T cells, suggesting that DMBG abrogated metabolic suppression in both cell types. DMBG treatment also shortened the amount of time it took for T cells suppressed by MDSCs to regain their capacity to respond to activation.

Methylglyoxal is known to react with the amino acid L-arginine, which is required for T cell activation. Investigating whether this interaction might play a role in the suppression of T cell activation, the researchers looked at T cells following the transfer of methylglyoxal from MDSCs, and found that L-arginine was completely reduced and that methylglyoxal-derived glycation products of L-arginine were increased, suggesting that methylglyoxal depletes L-arginine and may render L-arginine-containing proteins non-functional.

Looking at samples from patients with liver cancer, Baumann et al. found that MDSCs from tumors were stronger at suppressing CD8+ T cell proliferation than MDSCs from peritumoral tissue or blood. DMBG reversed the suppressive effect of all human M-MDSCs. PMN-MDSCs could not be sufficiently isolated for testing, however, those from blood did not show evidence of methylglyoxal accumulation or suppressive activity.

Finally, to determine whether the mechanism they had uncovered could be utilized to enhance immunotherapy, Baumann et al. tested a therapeutic OVA vaccine, DMBG treatment, and anti-PD-1 in mice with B16-OVA melanoma. None of the treatments were effective as monotherapies, however, the combination of the OVA vaccine with DMBG treatment reduced tumor growth, and the combination of DMBG treatment with anti-PD-1 strongly reduced tumor growth. Characterization of the tumor immune contexture showed that most of the MDSC-like cells showed evidence of methylglyoxal accumulation, displayed an inability to take up glucose, and suppressed CD8+ T cell proliferation when cultured ex vivo. In DMBG-treated mice, these effects were not observed.

Overall, Baumann et al. report that methylglyoxal is a metabolic marker that accumulates in MDSCs and greatly reduces metabolic functions. They also uncovered that MDSCs transfer methylglyoxal to T cells through direct cell contact, rendering T cells metabolically dysfunctional and temporarily “stunned” – unable to respond to T cell activation signals. This occurs because methylglyoxal depletes L-arginine and may affect the production of L-arginine-derived protein products. Neutralization of methylglyoxal using DMBG overcame MDSC-mediated T cell suppression and, in a murine tumor model, improved the efficacy of immunotherapies.

by Lauren Hitchings

Meet the researcher

This week, lead author Bastian Höchst answered our questions.

First author Tobias Baumann (left) and lead author Bastian Höchst (right)

What prompted you to tackle this research question?
The basic idea was that I always had problems with the idea that MDSCs are suppressive via NO, ROS and arginase, especially since other cells (type I macrophages, granulocytes,  hepatocytes) produce significantly more of these substances/enzymes and are not generally suppressive. Therefore, I assumed that there must be one or more mechanisms besides these factors.

What was the most surprising finding of this study for you?
The biggest surprise was the fact that there is a cytosolic exchange of substances, which also happens very quickly. We observed that this transport could happen within minutes, and that this exchange seemed to take place relatively often, at least between immune cells, with sometimes massive effects.

What was the coolest thing you’ve learned (about) recently outside of work?
The most interesting thing I have learned outside of work is that genes have a great influence on the behaviour and nature of children. Apparently, even behavioral traits are inherited in this way.

References:

Baumann T., Dunkel A., Schmid C., Schmitt S., Hiltensperger M., Lohr K., Laketa V., Donakonda S., Ahting U., Lorenz-Depiereux B., Heil J.E., Schredelseker J., Simeoni L., Fecher C., Körber N., Bauer T., Hüser N., Hartmann D., Laschinger M., Eyerich K., Eyerich S., Anton M., Streeter M., Wang T., Schraven B., Spiegel D., Assaad F., Misgeld T., Zischka H., Murray P.J., Heine A., Heikenwälder M., Korn T., Dawid C., Hofmann T., Knolle P.A., Höchst B. Regulatory myeloid cells paralyze T cells through cell-cell transfer of the metabolite methylglyoxal. Nat Immunol. 2020 May.

In the Spotlight...

Microbiota-Induced Type I Interferons Instruct a Poised Basal State of Dendritic Cells

Schaupp, Muth, and Rogell et al. showed that cDCs of mice lacking commensal microbiota had impaired anti-CD40-induced cytokine responses, reversible by bacterial recolonization. RNAseq, biochemical, and reporter assays showed that microbiota signaled pDCs via pattern recognition receptors to produce basal levels of IFNβ that induced type I IFN receptor-dependent tonic signaling in mature cDCs. This signaling instructed specific epigenomic and metabolic programs that enabled CD8+ T cell priming in a model system with cDC-expressed “self”-antigens. Anti-CD40 stimulation did not overcome impaired cDC response resulting from Type I IFN deficiency.

Contributed by Paula Hochman

Ectopic Tcf1 expression instills a stem-like program in exhausted CD8+ T cells to enhance viral and tumor immunity

Using Tcf1 transgenic mice, Shan and Hu et al. showed that ectopically expressed transcription factor Tcf1 skewed exhausted CD8+ T cells toward a stem-like phenotype (Tex-stem), which had enhanced proliferative capacity, persistence, and cytokine production, and reduced PD-1 expression. Tex-stem cells improved control of B16 tumors and LCMV chronic viral infection, and exhibited enhanced response to anti-PD-L1. Mechanistically, ectopic Tcf1 expression enhanced gene expression patterns established in WT Tcf1+ Tex-stem cells, but also led to expression of new genes via modulation of existing and novel chromatin accessibility sites.

Contributed by Anna Scherer

CLINICAL TRIAL: Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer

Lu et al. report the safety, feasibility, and efficacy of first-in-human phase I clinical trial of CRISPR Cas9 PD1-edited T cell infusion in patients with NSCLC. Editing frequency was low (~20%), and edited CD8+ T cells showed decreased PD-1 expression and an increase in IFNγ positivity. Sequencing showed limited off-target effect in the edited cells, and the treatment-related adverse events were of grades 1 and 2. Edited PD-1 gene and unique TCR clones were detectable in PBMCs and were persistent in a patient with stable disease. Among 12 treated patients, median PFS was 7.7 weeks and the best response was stable disease in 2 patients.

Contributed by Shishir Pant

PD-1 blockade-unresponsive human tumor-infiltrating CD8+ T cells are marked by loss of CD28 expression and rescued by IL-151

Using single-cell data of CD8+ TILs, Kim et al. show distinct expression patterns for CD28 in mouse and human tumors. In humans, CD28 expression by CD8+ T cells served as a marker for, but was functionally irrelevant to, anti-PD-1 antibody responsiveness. Human CD28-PD-1+CD8+ TILs show features of terminal exhaustion and high effector-like function, and were associated with poor prognosis and overall survival in anti-PD-1-treated patients with melanoma. IL-15 treatment ex vivo increased CD28-/CD28+ PD-1+CD8+ human TIL proliferation, and the combination of IL-15/IL-15Rα with anti-PD-1 enhanced antitumor response in a mouse tumor model.

Contributed by Shishir Pant

CCL5 mediates CD40-driven CD4+ T-cell tumor infiltration and immunity

To investigate the mechanism of agonist (ag) CD40-driven intratumoral CD4+ T cell chemotaxis, Huffman and Lin et al. evaluated the effects of checkpoint blockade (ICB) with or without agCD40 in a SQ model of pancreatic cancer, requiring both CD4+ and CD8+ T cells for tumor regression. 10X Genomics scRNA sequencing revealed that agCD40/ICB broadly increased chemokine CCL5 expression by intratumoral myeloid cells, compared to ICB alone, and tumor regression was abrogated by CCL5 knockout or anti-CCL5 treatment. CCL5 recruitment was selective for CD4+ T cells, highlighting a role in CD4+ T cell-mediated tumor chemotaxis.

Contributed by Katherine Turner

Commensal bacteria stimulate antitumor responses via T cell cross-reactivity

Bessell et al. identified an epitope expressed on commensal bacteria (SVY) with homology to a model tumor antigen (SIY) capable of eliciting cross-reactive T cell responses. Despite different TCR-pMHC affinities between the two epitopes, T cells expanded against SVY or SIY cross-reacted with the other and shared T cell clones. Mice exposed to commensal bacteria expressing SVY, which varied by commercial source (or in some experiments, by co-housing), had slower B16-SIY tumor growth and could generate stronger SVY-specific T cell responses with altered clonal composition. Adoptively transferred SVY-specific T cells slowed B16-SIY growth.

Contributed by Alex Najibi

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.