Weekly Digests
‹ Back to October

Understanding histone lactylation to enhance immunotherapy

October 23, 2024

Metabolic changes in T cells are often accompanied by changes in gene expression, functionality, and differentiation state. To better understand the connections between some of these changes, Raychaudhuri and Singh et al. recently explored how metabolic pathways that increase lactate production can lead to changes in lactylation – a histone modification of lysine residues – and uncovered how that contributed to the regulation of various changes in T cells following TCR activation.

To begin, Raychaudhuri and Singh et al. evaluated histone lactylation patterns in naive and effector CD8+ T cells, and found that following activation with anti-CD3 or a cognate antigen, intracellular and extracellular lactate accumulated, as did H3K18 lactylation (H3K18la) and H3K9 lactylation (H3K9la), particularly in cells expressing effector markers like granzyme B, TBET, and IFNγ. This pattern of increased lactylation in highly activated cells was evident in cultures of both human and murine T cells, a murine GvHD model, a murine tumor model, and a murine tumor model treated with immune checkpoint blockade (anti-PD-1 + anti-CTLA-4).

To better understand the role of H3K18la and H3K9la in transcriptional regulation, the researchers performed ChIPseq and ChromHMM analysis to classify genomic regions into various states based on patterns of histone post-translational modifications (hPTMs), including modifications that initiated transcription, marked active enhancer regions, associated with transcribing gene bodies, and repressed transcription. This analysis showed that H3K18la and H3K9la were enriched near transcription start sites (TSSs) and CpG islands, and were associated with other transcription-initiating hPTMs in promoter regions and in proximal and distal (but not intergenic) enhancer-like sequence-bearing regions, suggested that lactylation was involved in initiating transcription.

In naive T cells, H3K9la was enriched in promoter regions and was increased following T cell activation. H3K18la, on the other hand, was minimally enriched in naive T cells, but was enriched on promoter regions following activation. Both H3K18la and H3K9la were enriched in proximal enhancer-like regions following T cell activation, but only H3K18la was enriched in distal enhancer-like regions. Regions marked by lactylation peaks also showed higher occupancy of RNApol II, suggesting enhanced transcription.

Integrating RNAseq data with their CHIPseq data, the researchers found that in activated CD8+ T cells, H3K18la (alone or in combination with other hPTMs) marked genes that were crucial for T cell activation and effector molecules (Stat1, Cd28, Icos, Pdcd1, Pfkfb2, Fasl, Zap70, Gzmb, and Prf1), while H3K9la was enriched in the T cell activation gene Ifng and in genes associated with naive and memory T cells (Tcf7, Ccr7, and Batf3). Transcription factor (TF) binding motif enrichment analysis further showed that H3K18la-marked promoter regions were highly enriched in ETS binding motifs, while H3K9la-marked promoter regions were enriched for zinc finger binding motifs; both ETS and zinc finger TFs are associated with CD8+ T cell differentiation and functionality. In naive CD8+ T cells, H3K9la was enriched in genes encoding molecules involved in T cell quiescence (Bach2, Foxo1, and Klf2), and in memory T cells, H3K9la was enriched in genes associated with memory (Lef1, Ccr7, and Il7r). Both H3K18la and H3K9la were limited in exhausted CD8+ T cells, and did not show enrichment in exhaustion-associated genes.

Next, the researchers used gene set enrichment analysis (GSEA) and found that in activated CD8+ T cells, H3K18la was linked to increased JAK–STAT signaling, inflammatory responses, genes involved in glycolysis, and enzymes involved in glucose metabolism, while H3K9la was linked to increased OXPHOS, enzymes involved in OXPHOS, decreased TNF signaling, and decreased cytokine–cytokine receptor interaction pathways. In memory T cells, H3K9la was linked to inflammatory responses and fatty acid metabolism, and in naive T cells, it was linked to enzymes involved in mitochondrial metabolism. H3K18la was also linked to mitochondrial fission during CD8+ T cell activation, whereas H3K9la was linked to mitochondrial fusion in naive and memory CD8+ T cells. Together, these findings suggest that H3K9la supports sustained mitochondrial metabolism, while H3K18la mediates the shift towards glycolysis during CD8+ T cell activation.

Compared to naive and memory T cell subsets, which rely mostly on OXPHOS and fatty acid oxidation, activated CD8+ T cells rely mostly on glycolysis, which leads to an accumulation of lactate. To determine whether this metabolic shift contributes to increased lactylation, the researchers used an LDHA inhibitor to inhibit lactate production, and found that this also inhibited H3K18la and H3K9la, and shifted mitochondrial morphology from fission towards fusion. Similarly, blocking lactate export increased lactylation in activated T cells, but not in memory T cells, where glycolysis and lactate accumulation were less prevalent. Instead, inhibition of the electron transport chain, fatty acid oxidation, or ATP citrate lyase reduced H3K9la in memory T cells, where these metabolic pathways were more dominant. Administration of exogenous sodium-L-lactate (Na-La) increased H3K18la and H3K9la in naive and memory, but not activated CD8+ T cells, dependent on lactate transport into the cells. In naive cells, Na-La increased H3K9la at gene promoters associated with naiveness, and drove increased expression of those genes.

To test whether modulation of H3K18la and H3K9la levels could be utilized to modulate the functionality of activated CD8+ T cells, the researchers treated them with LDHA, thereby inhibiting lactate production and subsequent lactylation. This reduced H3K18la and H3K9la on activation-associated gene promoters and reduced expression of those genes, ultimately reducing production of cytotoxic molecules and the capacity for antigen-specific T cell-mediated killing. Similar effects were observed with inhibition of CBP/EP300 – a known histone lactylase. However, targeting HDAC1–HDAC3 – known histone delactylases – with the HDAC inhibitor MS275 increased H3K18la and H3K9la, increasing the expression of effector genes and the capacity for antigen-specific T cell-mediated killing. In a mouse tumor model, treatment with MS275 increased H3K18la and H3K9la in tumor- and tdLN-derived CD8+ T cells, increased intratumoral granzyme B+ effector CD8+ T cells, and reduced tumor growth. Inhibition of HNO1, a histone lactylase, had the opposite effect.

This study begins to unravel the complex relationship between histone lactylation and metabolic, transcriptional, and functional changes that occur in T cells, particularly in response to TCR activation. Importantly, it also establishes modulation of histone lactylation as a potential strategy for enhancing the functionality of CD8+ T cells in tumor settings, opening up new avenues of exploration in cancer immunotherapy.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, lead author Sangeeta Goswami answered our questions.

Current lab members involved in the study

What was the most surprising finding of this study for you?
One of the most surprising findings in our study was discovering how lactate, derived from different metabolic pathways, influenced specific histone modifications in various CD8+ T cell subsets. This raised an intriguing question: why do these metabolic sources selectively modify different histone residues? A major challenge we faced was that no specific writers and erasers of histone lactylation were known. So, we had to block multiple metabolic and epigenetic pathways to characterize the impact of histone lactylation in regulating T cell function. Additionally, our computational analysis showed that even though there were other histone modifications that regulated gene transcription, histone lactylation was critical for gene transcription in CD8+ T cells.

What is the outlook?
We expect our findings to shed light on how histone lactylation interacts with other histone modifications to regulate gene transcription in CD8+ T cells, which play an important role in fighting cancer and infection. Applying these insights to current clinical trials may also explain why some patients respond to immunotherapy while others don't, guiding new strategies to improve treatment outcomes.

What was the coolest thing you’ve learned (about) recently outside of work?
As a physician, scientist, mentor, and a mother, maintaining an optimum work-life balance is always an aspirational goal. However, playing each of these individual roles has taught me that they are interconnected, and that nothing brings greater joy than seeing the sense of happiness and pride in people I truly care about. For instance, seeing a therapeutic response in a patient, happiness in my mentees face on publishing a paper, or a sense of pride on my son’s face after his first orchestra concert.

References:

Raychaudhuri D, Singh P, Chakraborty B, Hennessey M, Tannir AJ, Byregowda S, Natarajan SM, Trujillo-Ocampo A, Im JS, Goswami S. Histone lactylation drives CD8+ T cell metabolism and function. Nat Immunol. 2024 Oct 7. 

In the Spotlight...

A comprehensive proteogenomic pipeline for neoantigen discovery to advance personalized cancer immunotherapy

Huber et al. developed a pipeline (Neodisc) integrating WGS/WES, RNAseq, and immunopeptidomics to predict and prioritize antigenic peptides derived from mutations, TSAs, TAAs, oncoviruses, or non-canonical transcripts for clinical use. Neodisc more efficiently ranked peptide candidates than existing approaches. LOH events impacting HLA alleles and other defects in the tumor antigen processing machinery could be analyzed to account for peptide presentation capacity, and results from multiple biopsies could be merged into one pipeline to study tumor heterogeneity.

Contributed by Morgan Janes

A prospective study of neoadjuvant pembrolizumab plus chemotherapy for resectable esophageal squamous cell carcinoma: The Keystone-001 trial

Shang, Xie, and Yu et al. report that neoadjuvant pembrolizumab plus chemotherapy in patients with resectable esophageal squamous cell carcinoma was well tolerated, without delays in subsequent surgery. The quality of life and nutritional status improved following neoadjuvant therapy, with a 2-year OS of 91%, and major pathological responses in 72% of patients. TRGC2+ NKT cell levels before treatment and their expansion after treatment correlated with response. TRGC2+ NKT cells increased cytotoxicity against patient-derived organoids ex vivo, and high TRGC2+ NKT cell infiltration was associated with better responses to immunotherapy in pan-cancer datasets.

Contributed by Shishir Pant

CAR-redirected natural killer T cells demonstrate superior antitumor activity to CAR-T cells through multimodal CD1d-dependent mechanisms

Zhou et al. showed that human CD19-targeted murine CAR-NKT and CAR-T cells exhibited comparable antitumor activity in vitro, but CAR-NKT cell treatment provided greater efficacy in multiple immunocompetent solid tumor models. CAR-NKT cell treatment altered the TME by reducing M2-like macrophages and activating DCs to promote responses and epitope spreading to endogenous T cells in a CD1d-dependent manner, complementing direct CAR-NKT cell activity. PD-1+Tim3+ exhausted CAR-NKT cells were detected in a model of high tumor burden. PD-1 blockade and α-GalCer vaccination boosted CAR-NKT cell antitumor activity.

Contributed by Paula Hochman

GLUT1 overexpression in CAR-T cells induces metabolic reprogramming and enhances potency

In this study, Guerrero and Klysz et al. stably overexpressed (OE) the glucose transporter GLUT1 in primary human CAR-T cells to determine if enhancing glucose availability would improve their function and antitumor potency. GLUT1OE in CAR-T cells enhanced glycolysis and oxidative phosphorylation, and was associated with decreased T cell exhaustion, increased Th17 differentiation, and increased glutathione-mediated resistance to ROS. In tumor-challenged mice, GLUT1OE CAR-T cells secreted more proinflammatory cytokines, and demonstrated superior tumor cell clearance and persistence compared to control CAR-T cells.

Contributed by Katherine Turner

Lipopolyplex-formulated mRNA cancer vaccine elicits strong neoantigen-specific T cell responses and antitumor activity

Predicted neoantigens from the CT26, MC38, and B16F10 mouse tumor models were encapsulated as mRNA in lipopolyplexes (LPPs). Injected s.c., the vaccine activated DCs and T cells, elicited neoantigen-specific and memory T cells, slowed tumor growth (dependent on CD8+ T cells), protected against tumor rechallenge, and synergized with anti-PD-1. 26/60 predicted neoantigens were immunogenic. Two human patients with advanced solid tumors were treated with a personalized LPP-mRNA vaccine (in combination with other therapies); treatment was safe, neoantigen-specific T cells were detected, and tumor outcomes were favorable.

Contributed by Alex Najibi

Overcoming tyrosine kinase inhibitor resistance in lung cancer brain metastasis with CTLA4 blockade

Fu and Zhao et al. demonstrated that tyrosine kinase inhibitors (TKI) of lung cancer brain mets (LCBM) showed bidirectional effects on T cells, augmenting T cell infiltration and impairing effector functions through CTLA-4 expression. Mechanistically, TKI treatment induced tumor cell secretion of HMGB1, which induced NF-κB activation and subsequent chemokine expression in macrophages, leading to increased T cell recruitment and upregulation of CTLA-4 expression in T cells. CTLA-4 blockade following Osimertinib significantly reduced LCBM burden, prolonged overall survival, and enhanced T cell antitumor activity in a LCBM murine model.

Contributed by Shishir Pant

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.