• HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
Saturday, November 22, 2025
BIOENGINEER.ORG
No Result
View All Result
  • Login
  • HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
        • Lecturer
        • PhD Studentship
        • Postdoc
        • Research Assistant
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
  • HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
        • Lecturer
        • PhD Studentship
        • Postdoc
        • Research Assistant
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
No Result
View All Result
Bioengineer.org
No Result
View All Result
Home NEWS Science News Cancer

Lactylation Marks Tumor Clusters, Predicts Glioblastoma Outcome

Bioengineer by Bioengineer
November 22, 2025
in Cancer
Reading Time: 4 mins read
0
Share on FacebookShare on TwitterShare on LinkedinShare on RedditShare on Telegram

Glioblastoma (GBM) stands out as the most malignant and aggressive form of adult brain cancer, notorious for its remarkable intratumoral heterogeneity and resistance to conventional therapies. Despite ongoing advancements in neuro-oncology, the prognosis for GBM patients remains grim, with survival rates stubbornly low. A groundbreaking study published in BMC Cancer in 2025 has illuminated a novel facet of GBM biology—histone lactylation—and its profound implications for tumor progression, immune evasion, and patient prognosis. Utilizing cutting-edge single-cell and spatial transcriptomics technologies, researchers have begun to unravel the complex cellular and molecular landscape shaped by lactylation within GBM tumors.

Histone lactylation is an emerging epigenetic modification that links metabolic changes, particularly in the tumor microenvironment, to gene expression alterations driving cancer development. This study leverages a multi-omics approach, integrating bulk RNA sequencing, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics, to dissect the role of lactylation in GBM at unprecedented resolution. By probing datasets from GEO and TCGA, the team identified lactylation-related genes that are markedly upregulated in GBM tissues and are associated with immunosuppressive microenvironments and poor clinical outcomes.

A key finding centers around the discovery of distinct malignant tumor cell subpopulations exhibiting high levels of lactylation, which reside predominantly within hypoxic regions of the tumor core. These hypoxic niches are well-known for fostering aggressive tumor phenotypes that evade immune surveillance. Single-cell analyses revealed that these lactylated clusters undergo profound metabolic reprogramming, tailoring their gene expression to survive and thrive under oxygen-deprived conditions, while concurrently orchestrating mechanisms to suppress the surrounding immune response.

Spatial transcriptomics added another critical dimension to the findings by mapping the precise localization of these lactylated tumor cells within the heterogeneous tumor architecture. In particular, cells expressing high levels of S100A6, a gene intimately linked to lactylation, were found concentrated in aggressive tumor regions notorious for rapid proliferation and invasion. This spatial information underscores the functional heterogeneity within GBM and provides a tangible target for therapeutic interventions.

To translate these molecular insights into clinical practice, the researchers developed a prognostic risk model based on nine lactylation-associated genes. Using LASSO-Cox regression—a powerful statistical method for feature selection—they stratified GBM patients into distinct high- and low-risk groups. Strikingly, this model demonstrated impressive predictive accuracy with area under the curve (AUC) values ranging from 0.77 to 0.87, suggesting its potential utility as a robust biomarker panel for patient prognosis and treatment stratification.

The compelling prognostic value of the lactylation signature is further supported by experimental validation. In vitro functional assays targeting S100A6 demonstrated that silencing this gene significantly impaired GBM cell proliferation, migration, and invasion, highlighting its pivotal role in maintaining tumor aggressiveness. These findings position S100A6 not merely as a biomarker but as a potential therapeutic target for disrupting lactylation-driven malignant programs.

Underpinning these discoveries is the innovative application of SCENIC transcriptional network inference and CellChat intercellular communication modeling. These computational tools enabled the authors to uncover regulatory networks and cell-cell interactions modulated by lactylation, providing mechanistic insights into how tumor cells rewire signaling pathways to foster immune suppression and metabolic adaptation in GBM.

Pseudotime trajectory analyses further delineated the dynamic states of tumor cell populations, tracing the evolutionary paths from less aggressive to more malignant lactylated states. This temporal framework enriches our understanding of tumor progression and highlights critical junctures where therapeutic interventions might be most effective.

The study also sheds light on the tumor immune microenvironment, revealing that lactylation-associated clusters contribute to the establishment of immunosuppressive niches. This finding dovetails with accumulating evidence that metabolic reprogramming in tumors orchestrates immune evasion, a major challenge for immunotherapies in GBM.

Moreover, the emergence of lactylation as a key metabolic-epigenetic axis opens avenues for novel therapeutic strategies. Targeting enzymes responsible for lactylation or the downstream effectors, such as S100A6, could potentially disrupt malignant metabolic circuits, sensitize tumors to immune attack, or enhance the efficacy of existing treatments.

Beyond its immediate clinical relevance, this research marks a significant advance in cancer biology by employing integrated single-cell and spatial transcriptomics to parse tumor complexity. This multidimensional profiling affords a holistic view of cellular heterogeneity, spatial organization, and functional states within tumors—an approach likely to become foundational in precision oncology.

Despite these promising findings, challenges remain. Validation of the prognostic model and therapeutic targets in larger, independent patient cohorts and in vivo models will be essential to confirm their utility. Additionally, translating knowledge of lactylation into safe and effective clinical interventions will require comprehensive understanding of the broader systemic effects of modulating this epigenetic mark.

Nevertheless, this study underscores the transformative potential of marrying metabolic insights with high-resolution transcriptomic technologies to redefine our understanding of glioblastoma. By pinpointing lactylation as a central player in tumor cell clustering, metabolic adaptation, and immune modulation, it opens a new chapter in the fight against one of the deadliest brain cancers.

In summary, this pioneering work reveals that lactylation is more than a metabolic footnote in glioblastoma biology; it is a defining feature of tumor heterogeneity and aggressiveness. The identification of lactylation-enriched tumor cell clusters, spatially anchored in hypoxic niches and regulated by signatures including S100A6, provides a powerful prognostic tool and therapeutic target. This research paves the way for the development of lactylation-focused strategies that could revolutionize glioblastoma treatment and improve outcomes for patients facing this devastating disease.

Subject of Research: Metabolic reprogramming through histone lactylation in glioblastoma, its association with tumor heterogeneity, immune evasion, and prognosis.

Article Title: Single-cell and spatial transcriptomics reveal lactylation-associated tumor cell clusters and define a prognostic risk model in glioblastoma

Article References:
Han, R., Chi, G., Sun, D. et al. Single-cell and spatial transcriptomics reveal lactylation-associated tumor cell clusters and define a prognostic risk model in glioblastoma. BMC Cancer (2025). https://doi.org/10.1186/s12885-025-15291-6

Image Credits: Scienmag.com

DOI: https://doi.org/10.1186/s12885-025-15291-6

Tags: epigenetic modifications and cancerglioblastoma researchhistone lactylation in cancerimmune evasion in glioblastomaIntratumoral Heterogeneity in GBMlactylation-related genes in cancermetabolic changes in brain tumorsmulti-omics approach in oncologyprognosis of glioblastoma patientssingle-cell transcriptomics in glioblastomaspatial transcriptomics and tumor analysistumor microenvironment and gene expression

Tags: İşte 5 adet uygun etiket: **glioblastoma laktilasyonumetabolik epigenetik** **Açıklama:** 1. **glioblastoma laktilasyonu:** Doğrudan makalenin ana konusunu (laktilasyonun GBM'deki rolü)prognostik belirteçlertek hücreli transkriptomiktümör heterojenitesi
Share12Tweet8Share2ShareShareShare2

Related Posts

Hope, Well-Being Shape Elderly Cancer Patients’ Quality

November 22, 2025

Urinary microRNA Differentiates Bladder Cancer Types

November 22, 2025

Immune Checkpoint Inhibitors Tested in Triple-Negative Breast Cancer

November 22, 2025

Plasma Sequencing Advances NSCLC Diagnosis, Treatment

November 22, 2025

POPULAR NEWS

  • New Research Unveils the Pathway for CEOs to Achieve Social Media Stardom

    New Research Unveils the Pathway for CEOs to Achieve Social Media Stardom

    202 shares
    Share 81 Tweet 51
  • Scientists Uncover Chameleon’s Telephone-Cord-Like Optic Nerves, A Feature Missed by Aristotle and Newton

    119 shares
    Share 48 Tweet 30
  • Neurological Impacts of COVID and MIS-C in Children

    92 shares
    Share 37 Tweet 23
  • ESMO 2025: mRNA COVID Vaccines Enhance Efficacy of Cancer Immunotherapy

    211 shares
    Share 84 Tweet 53

About

We bring you the latest biotechnology news from best research centers and universities around the world. Check our website.

Follow us

Recent News

Hope, Well-Being Shape Elderly Cancer Patients’ Quality

Mouse Serum Promotes Follicular Cavity Expansion Across Estrous Cycles

Body Fat and Brain: Interconnected Health Dynamics

Subscribe to Blog via Email

Enter your email address to subscribe to this blog and receive notifications of new posts by email.

Join 69 other subscribers
  • Contact Us

Bioengineer.org © Copyright 2023 All Rights Reserved.

Welcome Back!

Login to your account below

Forgotten Password?

Retrieve your password

Please enter your username or email address to reset your password.

Log In
No Result
View All Result
  • Homepages
    • Home Page 1
    • Home Page 2
  • News
  • National
  • Business
  • Health
  • Lifestyle
  • Science

Bioengineer.org © Copyright 2023 All Rights Reserved.