• HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
Sunday, May 25, 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

Unraveling Tumor Microenvironment Dynamics: How Long Non-Coding RNAs Shape Cancer Progression

Bioengineer by Bioengineer
April 24, 2025
in Cancer
Reading Time: 4 mins read
0
ADVERTISEMENT
Share on FacebookShare on TwitterShare on LinkedinShare on RedditShare on Telegram

Tumor Microenvironment Dynamics: The Regulatory Influence of Long Non-coding RNAs

Long non-coding RNAs (lncRNAs) are emerging as pivotal regulators within the tumor microenvironment (TME), reshaping our understanding of cancer biology and opening new avenues for therapeutic intervention. Defined as RNA molecules longer than 200 nucleotides that do not encode proteins, lncRNAs were once deemed transcriptional noise. However, recent discoveries have revealed their profound influence on gene regulation, intercellular signaling, and the dynamic interplay between tumor cells and the surrounding stromal and immune cells. These molecules intricately modulate key processes including immune evasion, angiogenesis, metastasis, and resistance to therapy, positioning lncRNAs at the forefront of cancer research innovation.

The tumor microenvironment is a highly complex ecosystem comprising heterogeneous populations of cancer cells, fibroblasts, immune infiltrates, extracellular matrix (ECM) components, and a myriad of soluble factors such as cytokines and growth factors. Within this network, lncRNAs act as master regulators, orchestrating communication between diverse cell types and modulating the extracellular milieu to favor tumor progression. They do so through multifaceted mechanisms that operate at transcriptional, post-transcriptional, and epigenetic levels, influencing chromatin remodeling, mRNA stability, and protein translation. This multilayered regulatory capacity endows lncRNAs with the ability to fine-tune molecular pathways critical for tumor survival and expansion.

One of the critical elements of lncRNA function in the TME is their role in controlling stromal-tumor crosstalk, particularly through tumor-associated fibroblasts (TAFs). These fibroblasts, reprogrammed by tumor-derived signals, contribute to ECM remodeling, immune modulation, and secretion of growth factors. Specific lncRNAs, such as LOC100506114 in oral squamous cell carcinoma, have been shown to induce fibroblast activation, enhancing their pro-tumorigenic potential. Furthermore, overexpression of MALAT1 in TAFs correlates with increased invasiveness and migratory capacity of adjacent tumor cells, illustrating the direct influence of lncRNAs on cellular behavior within the stroma.

Beyond modulating fibroblast activity, lncRNAs profoundly impact immune cell dynamics within the TME, often creating an immunosuppressive niche that allows tumors to evade immune surveillance. Through the regulation of immune checkpoints and cytokine production, lncRNAs such as HISLA facilitate metabolic reprogramming of tumor-associated macrophages (TAMs), promoting glycolysis and anti-apoptotic pathways in cancer cells. Additionally, lncRNAs NEAT1 and MALAT1 have been implicated in impairing T cell function by modulating immune checkpoint molecules, thus attenuating the anti-tumor immune response and fostering an environment conducive to tumor persistence.

Angiogenesis, the formation of new blood vessels, is indispensable for tumor growth and metastasis as it ensures an adequate supply of oxygen and nutrients. Intriguingly, lncRNAs regulate angiogenic signaling cascades within both tumor and endothelial cells. The lncRNA NR2F1-AS1, for instance, correlates with endothelial markers CD31 and CD34 in breast cancer, directly facilitating vascular sprouting. Similarly, PVT1 promotes vascular endothelial growth factor A (VEGFA) expression in gastric cancer, thus intensifying the angiogenic drive. Therapeutic targeting of these lncRNAs could disrupt the vascular network that sustains tumors, underscoring their potential in anti-angiogenic strategies.

Intercellular communication in the TME is further complicated by the release and uptake of extracellular vesicles such as exosomes, which shuttle bioactive molecules including lncRNAs between cells. Exosomal lncRNAs exemplify a sophisticated mechanism through which tumors manipulate their environment. For example, the transfer of the lncRNA CRNDE from TAMs to gastric cancer cells via exosomes facilitates degradation of the tumor suppressor PTEN, thereby augmenting tumor cell survival and chemoresistance. This form of horizontal lncRNA transfer exemplifies how cancer cells can exploit the microenvironment to promote selective advantages while evading therapeutic pressures.

Resistance to conventional therapies remains a formidable obstacle in cancer treatment. lncRNAs are now recognized as pivotal in mediating both intrinsic and acquired resistance. For instance, lncRNA DNM3OS, upregulated in esophageal cancer-associated fibroblasts, enhances DNA damage response mechanisms, rendering tumor cells more resistant to radiotherapy. Moreover, lncRNAs can modulate the expression of drug transporters, anti-apoptotic factors, and signaling pathways underpinning therapy escape, highlighting their critical roles in treatment failure and cancer recurrence.

The clinical implications of these discoveries extend beyond mechanistic insights, as lncRNAs possess significant potential as diagnostic and prognostic biomarkers. Their tissue-specific expression and remarkable stability in bodily fluids make them attractive candidates for non-invasive cancer detection assays. Circulating exosomal lncRNAs, in particular, offer a dynamic snapshot of tumor status and could revolutionize early detection and real-time monitoring of therapeutic responses, moving oncology towards more personalized medicine paradigms.

Despite these opportunities, targeting lncRNAs therapeutically presents considerable challenges. Their diverse modes of action, context-dependent functions, and structural complexity necessitate innovative strategies for effective modulation. Current approaches include antisense oligonucleotides (ASOs), RNA interference (RNAi), and CRISPR-Cas based gene editing, each requiring precise delivery systems to the tumor locale. Nanotechnology advancements provide promising vectors for such delivery, potentially overcoming the barriers of specificity and minimizing off-target effects that have hindered broader clinical translation.

Ongoing research continues to unravel the breadth of lncRNA functions within the TME, emphasizing their role in modulating key signaling pathways such as epithelial-mesenchymal transition (EMT), metabolic reprogramming, and cancer stem cell maintenance. By influencing these pivotal processes, lncRNAs effectively shape tumor aggressiveness and metastatic potential. The integration of multi-omics and single-cell technologies promises to elucidate the contextual dependency of lncRNA activities, informing the development of more sophisticated and tailored therapeutic interventions.

As the oncology field embraces these insights, the translation of lncRNA-based therapies from bench to bedside remains a critical frontier. Combining lncRNA targeting with established treatments such as chemotherapy, radiotherapy, and immunotherapy may enhance therapeutic outcomes by dismantling tumor-protective microenvironmental barriers. This multifaceted approach reflects a new era in cancer management, wherein understanding and manipulating the TME at the RNA regulatory level could significantly alter disease trajectories.

Ultimately, the expanding knowledge of lncRNAs within the tumor microenvironment heralds a paradigm shift in cancer biology. These molecules not only serve as regulatory hubs orchestrating tumor progression but also represent promising clinical targets and biomarkers. Continued interdisciplinary efforts integrating molecular biology, genomics, bioinformatics, and nanomedicine will be essential to harness their full potential, paving the way for innovative, effective, and personalized cancer therapies.

Subject of Research: Long non-coding RNAs in tumor microenvironment regulation and cancer progression

Article Title: Tumor Microenvironment Dynamics: The Regulatory Influence of Long Non-coding RNAs

News Publication Date: 22-Feb-2025

Web References:

Gene Expression Journal
DOI: 10.14218/GE.2024.00069

Image Credits: Ilgiz Gareev, Ozal Beylerli

Keywords: Long noncoding RNA, Primary tumors, Tumor cells, Molecular targets, Tumor microenvironments

Tags: cancer progression regulatory mechanismsepigenetic roles of lncRNAsextracellular matrix and cancergene regulation by lncRNAsintercellular signaling in tumorslncRNAs and angiogenesislncRNAs and immune evasionlncRNAs and therapy resistancelncRNAs in metastasislong non-coding RNAs in cancertherapeutic interventions targeting lncRNAstumor microenvironment dynamics

Tags: angiogenesis regulationCancer immune evasionextracellular vesicles in cancerLong non-coding RNAstumor microenvironment dynamics
Share12Tweet8Share2ShareShareShare2

Related Posts

MicroRNAs Driving Colorectal Cancer: Quick Review

MicroRNAs Driving Colorectal Cancer: Quick Review

May 24, 2025
blank

European Regulation Proposed to Prevent Transmission of Cancer-Linked Genetic Mutations via Sperm Donation

May 23, 2025

Machine Learning Predicts Breast Cancer Outcomes

May 23, 2025

Research Spotlight: Novel Therapy Blocks Glioblastoma’s Immune System Hijack

May 23, 2025

POPULAR NEWS

  • Effects of a natural ingredients-based intervention targeting the hallmarks of aging on epigenetic clocks, physical function, and body composition: a single-arm clinical trial

    Natural Supplement Shows Potential to Slow Biological Aging and Enhance Muscle Strength

    92 shares
    Share 37 Tweet 23
  • Analysis of Research Grant Terminations at the National Institutes of Health

    80 shares
    Share 32 Tweet 20
  • Health Octo Tool Links Personalized Health, Aging Rate

    68 shares
    Share 27 Tweet 17
  • Universe Fades Faster Than Expected—Yet Still Over Vast Timescales

    55 shares
    Share 22 Tweet 14

About

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

Follow us

Recent News

Analysis of 400,000 Women Validates BRCA Variant Classification

NSUN2 Drives Glycolysis, Immune Evasion in Kidney Cancer

Unequal Testing Skews Vaccine Effectiveness Estimates

  • 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.