Showing posts with label gene expression. Show all posts
Showing posts with label gene expression. Show all posts

Tuesday, August 12, 2025

How RNA Regulates Metabolic Stress

  

Role of RNA in Metabolic Stress (Image by ©TheRNABlog).


The RNA-Centric Nexus of Metabolic Stress: From Transcriptional Reprogramming to Epitranscriptomic Control

The Landscape of Metabolic Stress

The ability of a cell to sense and adapt to its metabolic environment is fundamental to life. When homeostatic balance is disrupted, cells enter a state of metabolic stress, a condition that encompasses a wide spectrum of physiological and pathological states. Understanding the multifaceted nature of metabolic stress is crucial, as it represents the upstream trigger for a cascade of sophisticated molecular responses, many of which are orchestrated by ribonucleic acid (RNA). The definition of metabolic stress is context-dependent, varying across biological disciplines, yet these different perspectives converge on a common set of core cellular perturbations that are ultimately interpreted by the cell's genetic and regulatory machinery.

Defining Metabolic Stress: A Context-Dependent Phenomenon

From a physiological standpoint, particularly in the field of exercise science, metabolic stress is defined as a process occurring in response to low energy availability during intense physical activity. This state is characterized by the accumulation of specific metabolites within muscle cells, including lactate, inorganic phosphate (Pi), and hydrogen ions (H+).  This accumulation is a direct result of elevated rates of ATP hydrolysis and glycolytic flux required to fuel muscle contractions.  The physiological consequences are significant and include the stimulation of anabolic hormonal release (e.g., growth hormone), increased production of reactive oxygen species (ROS), the induction of a local hypoxic environment due to the compression of blood vessels, and cell swelling.  These factors, driven by metabolite buildup, are considered primary mechanisms that stimulate muscle adaptations, including hypertrophy. In a broader cellular and pathophysiological context, the concept of metabolic stress extends beyond exercise-induced changes. It represents a more general disruption of cellular homeostasis triggered by a range of stimuli, including nutrient imbalance—both deprivation and excess—as well as hypoxia and oxidative stress.  This definition is particularly relevant to the study of chronic diseases. In neurodegenerative disorders such as Alzheimer's disease (AD), metabolic stress manifests as a complex interplay of neuroinflammation, insulin resistance, and endoplasmic reticulum (ER) stress within the brain.  Similarly, in metabolic disorders like type 2 diabetes (T2DM), chronic exposure to high levels of glucose and lipids (glucolipotoxicity) induces a state of metabolic stress in tissues like the pancreas, liver, and muscle, driving cellular dysfunction and pathology.  These varied definitions are not contradictory; rather, they describe different manifestations of a fundamental problem. High-level physiological events, whether a bout of resistance training or the progression of a chronic disease, induce a common, limited set of core cellular states. It is these core states that are directly sensed by the RNA regulatory machinery, revealing an elegant modularity in the cell's design. The cell does not require a unique response system for every conceivable external threat but instead possesses a robust, integrated set of responses to a few core internal challenges.

The Core Cellular Stressors: Proximate Triggers of the RNA Response

The diverse physiological phenomena classified as metabolic stress converge upon a handful of fundamental cellular challenges that serve as the proximate triggers for the vast network of RNA-mediated responses.

Nutrient and Energy Imbalance: This is a primary stressor that can manifest as either nutrient deprivation or nutrient excess. During deprivation, such as caloric restriction or fasting, cells activate key energy sensors like AMP-activated protein kinase (AMPK) and switch to more efficient catabolic pathways like mitochondrial oxidative phosphorylation.  While this adaptation maximizes ATP production from limited resources, it can paradoxically increase the generation of mitochondrial ROS.  Conversely, nutrient excess, a hallmark of modern metabolic diseases like obesity and T2DM, can overwhelm the cell's metabolic capacity, leading to ROS production that surpasses antioxidant defenses, thereby inducing a state of stress. Hypoxia: This condition of insufficient oxygen availability can be acute and physiological, as seen in exercising muscle, or chronic and pathological, as found in the microenvironment of solid tumors or in ischemic tissues following a stroke or heart attack.  Hypoxia is a potent activator of a master transcriptional program mediated by Hypoxia-Inducible Factors (HIFs), which orchestrate a cellular shift toward anaerobic metabolism and promote angiogenesis to restore oxygen supply. Oxidative Stress: This state arises from an imbalance between the production of ROS and the cell's antioxidant capacity.  Mitochondria and enzymes like NADPH oxidases (NOX) are major sources of ROS.  While physiological levels of ROS function as critical signaling molecules, excessive ROS are highly destructive, causing damage to all major classes of biological macromolecules, including lipids, proteins, and, critically, nucleic acids like DNA and RNA.  This damage is a key contributor to the aging process and the pathogenesis of numerous diseases. Endoplasmic Reticulum (ER) Stress: The ER is responsible for folding a significant portion of the cell's proteins. When the demand for protein folding exceeds the ER's capacity, unfolded or misfolded proteins accumulate, triggering a signaling cascade known as the Unfolded Protein Response (UPR).  ER stress is intimately linked with metabolic status, as nutrient fluctuations and oxidative stress can disrupt ER function. It is a central component of the metabolic stress observed in diseases like AD and T2DM, where it contributes to insulin resistance and inflammation.

The RNA Repertoire: An Overview of Key Molecular Players

To orchestrate the complex cellular response to metabolic stress, the cell deploys a diverse and versatile repertoire of RNA molecules. These range from the canonical players of the central dogma to a vast and intricate network of non-coding RNAs that function as regulators. An understanding of this cast of characters is essential to appreciate the multi-layered nature of the RNA-centric stress response.

The Central Dogma Players: More Than Just Messengers and Builders

The traditional roles of messenger RNA (mRNA), ribosomal RNA (rRNA), and transfer RNA (tRNA) as components of the protein synthesis machinery are well-established. However, under conditions of metabolic stress, these molecules transcend their housekeeping functions to become active participants in the regulatory network.

     Messenger RNA (mRNA): As the template for protein synthesis, mRNA carries genetic information from the nucleus to the ribosome.  The expression level of any given protein is a function of not only the transcription rate of its gene but also the stability and translational efficiency of its mRNA. These latter two properties are critical, dynamic regulatory nodes that are heavily modulated during metabolic stress.

     Ribosomal RNA (rRNA): Constituting the bulk of cellular RNA, rRNAs are the structural and catalytic heart of the ribosome, the cellular machine responsible for translation.  The catalytic activity of rRNA in forming peptide bonds marks it as a ribozyme, an RNA molecule with enzymatic function.  Far from being inert scaffolds, rRNAs are direct targets of oxidative damage during stress, which can impair ribosome function and contribute to the overall decline in protein synthesis.

     Transfer RNA (tRNA): These small, highly structured RNAs act as the adaptors that decipher the genetic code on mRNA and deliver the correct amino acids to the growing polypeptide chain.  tRNAs are subject to extensive chemical modifications that are vital for their stability and decoding accuracy. During metabolic stress, these modification patterns can be altered, and, remarkably, tRNAs can be specifically cleaved by enzymes to generate smaller tRNA-derived fragments (tRFs). These fragments are not merely degradation products but are emerging as a new class of regulatory small RNAs that can inhibit translation and participate in stress signaling pathways.  This functional plasticity, where core components of the translation machinery are repurposed into signaling molecules, represents a sophisticated layer of cellular adaptation, allowing the cell to generate new regulatory outputs from existing molecular pools without the need forde novo gene expression.

The Non-Coding Universe: A Symphony of Regulation

     The discovery that less than 2% of the human genome encodes proteins has revolutionized molecular biology, revealing that the majority of the genome is transcribed into non-coding RNAs (ncRNAs).  These molecules, which function directly as RNAs without being translated into protein, form a complex regulatory web that governs nearly every aspect of cell biology, and they are central to the metabolic stress response. Small ncRNAs: This category includes several classes of short RNA molecules.

     MicroRNAs (miRNAs): These are short (~22 nucleotide) RNAs that act as post-transcriptional repressors. They are loaded into an Argonaute protein complex and guide it to target mRNAs, typically through partial complementarity in the 3' untranslated region (UTR), leading to mRNA degradation or translational inhibition.  miRNAs are critical for fine-tuning entire gene networks and are key players in orchestrating cellular responses to metabolic challenges.

     Small nucleolar RNAs (snoRNAs): Found in the nucleolus, the primary role of snoRNAs is to guide the chemical modification (e.g., methylation and pseudouridylation) of rRNAs and other RNAs, a process essential for correct ribosome assembly and function.  Like tRNAs, snoRNAs are also being redefined in the context of stress, where they can be processed into smaller fragments with distinct regulatory functions and are implicated in the response to lipotoxicity and oxidative stress.

     Long non-coding RNAs (lncRNAs): This is a large and heterogeneous class of ncRNAs defined by a length greater than 200 nucleotides.  Their functional mechanisms are remarkably diverse; they can act as signals, decoys for miRNAs or proteins, guides that direct chromatin-modifying complexes to specific DNA loci, or scaffolds that assemble protein complexes.  A growing number of lncRNAs are specifically induced by metabolic stressors and have been identified as pivotal regulators in the pathogenesis of metabolic diseases.

The following table provides a summary of these key RNA classes and their dual roles in both homeostasis and metabolic stress.

 

RNA Class

Primary Function (Homeostasis)

Role/Fate in Metabolic Stress

mRNA

Protein-coding template

Altered stability and translational efficiency; direct oxidative damage

rRNA

Structural and catalytic core of the ribosome

Oxidative damage leading to impaired ribosome function; altered transcription

tRNA

Adaptor for amino acid delivery during translation

Cleavage into regulatory tRNA-derived fragments (tRFs); altered modification patterns

miRNA

Post-transcriptional gene silencing; fine-tuning of gene networks

Stress-specific expression (e.g., "hypoxamirs"); regulation of metabolic and apoptotic pathways

lncRNA

Diverse regulation (scaffold, guide, decoy, signal)

Stress-induced expression; epigenetic regulation; miRNA sponging; control of stress response pathways

snoRNA

Guidance of rRNA/snRNA chemical modification

Processing into smaller regulatory fragments; involvement in oxidative stress response

 

Transcriptional Reprogramming: The First Line of Genomic Defense

The most fundamental and enduring response to sustained metabolic stress is a profound reprogramming of the cell's transcriptional landscape. This process involves altering the expression of thousands of genes to suppress growth-related programs and activate protective and adaptive pathways. This global shift is orchestrated by a combination of stress-responsive transcription factors and dynamic changes in the chromatin environment, which itself acts as a sensor of the cell's metabolic state.

Orchestration by Stress-Responsive Transcription Factors (TFs)

Transcription factors are proteins that bind to specific DNA sequences to control the rate of transcription, thereby serving as the primary actuators of the gene expression response to cellular signals.  During metabolic stress, several key TFs are activated to mount a coordinated defense.

     Hypoxia-Inducible Factors (HIFs): In response to low oxygen, the HIF-1α subunit is stabilized, allowing it to dimerize with the constitutive HIF-1β subunit. This complex then translocates to the nucleus, where it binds to DNA sequences known as Hypoxia Response Elements (HREs) in the promoters of target genes.  This activates a broad transcriptional program that promotes adaptation to hypoxia, including the upregulation of genes involved in anaerobic glycolysis, angiogenesis (e.g.,VEGF), and erythropoiesis.

     Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2): Nrf2 is the master regulator of the antioxidant response.  Under homeostatic conditions, it is sequestered in the cytoplasm and targeted for degradation by the protein KEAP1. Oxidative stress disrupts this interaction, allowing Nrf2 to accumulate in the nucleus and activate the transcription of a battery of cytoprotective genes that contain Antioxidant Response Elements (AREs) in their promoters.  This response bolsters the cell's antioxidant defenses and detoxification capacity. Notably, this pathway is linked to metabolic sensing, as the energy sensor AMPK is required for the maximal transcriptional activation of Nrf2 targets.

     Activating Transcription Factor 4 (ATF4): As a central effector of the Integrated Stress Response (ISR), ATF4 is primarily regulated at the level of translation (discussed in Section 4.1). However, once synthesized, it functions as a potent transcription factor. ATF4 drives the expression of genes crucial for adapting to stress, including those involved in amino acid biosynthesis and transport, antioxidant responses, and ER chaperones. If the stress is severe and prolonged, the ATF4 program can switch from pro-survival to pro-apoptotic, initiating cell death.

 Chromatin as a Metabolic Sensor

The accessibility of genes for transcription is governed by the physical state of chromatin, the complex of DNA and histone proteins. This landscape is dynamically regulated by a series of post-translational modifications on histone tails and chemical modifications on DNA itself.  A profound link between metabolism and gene expression arises from the fact that the enzymes responsible for writing and erasing these epigenetic marks use key intermediary metabolites as essential substrates and cofactors.

This direct biochemical linkage means that the cell's metabolic state is translated into its epigenetic state, influencing its transcriptional potential. Key examples include:

     Acetylation: Histone acetylation, a mark generally associated with active transcription, is catalyzed by histone acetyltransferases (HATs) that use acetyl-CoA as the acetyl group donor. Cellular levels of acetyl-CoA are directly dependent on the metabolism of glucose, fatty acids, and acetate, thus linking nutrient availability directly to gene activation potential.

     Methylation: Histone and DNA methylation reactions, which can be either activating or repressive depending on the site, are catalyzed by methyltransferases that use S-adenosylmethionine (SAM) as the universal methyl donor. The SAM pool is maintained by the methionine cycle and one-carbon metabolism, pathways that are sensitive to the availability of specific amino acids and vitamins.

     Demethylation: A large class of demethylases, including the TET enzymes that hydroxylate methyl-cytosine in DNA and the JmjC-domain histone demethylases, are α-ketoglutarate-dependent dioxygenases. They require α-ketoglutarate (αKG), a central intermediate of the TCA cycle, as a co-substrate.  Consequently, their activity is sensitive to the balance between glycolysis and oxidative phosphorylation. Furthermore, they are competitively inhibited by metabolites like succinate, fumarate, and the oncometabolite D-2-hydroxyglutarate (D-2HG), which accumulate in certain metabolic diseases and cancers, leading to widespread hypermethylation and altered gene expression.

This intricate connection establishes a powerful feedback mechanism. The cell's metabolic state alters the availability of these key metabolites, which in turn modulates the activity of chromatin-modifying enzymes. The resulting changes in the chromatin landscape then regulate the expression of genes, including those encoding metabolic enzymes, thus reinforcing the initial metabolic state. This self-perpetuating loop can explain the stability of pathological metabolic states, such as the Warburg effect in cancer, and highlights the challenge in reversing them. Breaking this cycle may require therapeutic strategies that target both metabolic and epigenetic components simultaneously.

Global Transcriptomic Shifts in Response to Stress

The advent of high-throughput technologies like microarrays and RNA-sequencing has enabled an unbiased, genome-wide view of the transcriptional response to stress.  These transcriptomic studies have revealed that diverse stressors induce a common, coordinated program of gene expression changes. This typically involves the rapid induction of cytoprotective genes—such as molecular chaperones, antioxidant enzymes, and DNA repair factors—coupled with the widespread repression of genes associated with cell growth, proliferation, and anabolic metabolism. In the context of human disease, transcriptomic analyses of postmortem brain tissue from individuals with major depressive disorder (MDD) and post-traumatic stress disorder (PTSD)—conditions strongly linked to chronic stress—have uncovered significant dysregulation of gene networks. These networks are involved in synaptic function, neurotrophic factor signaling (e.g., BDNF), and the glucocorticoid response system.  These studies have also revealed striking sex-specific differences in the transcriptomic signatures of these disorders, suggesting that males and females may utilize different molecular pathways in their response to chronic stress.  The increasing use of single-cell and single-nucleus RNA-sequencing (scRNA-seq and snRNA-seq) is providing unprecedented resolution, allowing researchers to dissect the cell-type-specific transcriptional responses within complex tissues, further refining our understanding of how metabolic stress impacts organismal health.

Post-Transcriptional Regulation: Fine-Tuning the Stress Response

While transcriptional reprogramming establishes a new, long-term cellular state, it is a relatively slow process. To survive acute, transient stress, cells rely on a suite of faster, more dynamic post-transcriptional regulatory mechanisms. These processes act on RNA transcripts after they have been synthesized, controlling their translation, localization, and stability. This allows for a rapid and reversible modulation of gene expression, providing an immediate defense that buys time for the more profound transcriptional changes to take effect. This temporal layering of responses is a hallmark of a sophisticated and robust cellular defense system. An initial, rapid response based on post-transcriptional controls stabilizes the cell and mitigates immediate damage, while the slower, more comprehensive transcriptional program reconfigures the cell for long-term adaptation or survival.

Mechanism

Key Molecular Players

Primary Stress Triggers

Typical Response Timescale & Outcome

Translational Control via ISR

eIF2α kinases (PERK, GCN2, HRI, PKR), ATF4

ER stress, amino acid deprivation, oxidative stress, viral infection

Minutes; Global translation halt & selective translation of stress mRNAs

RNA Sequestration

G3BP1, TIA-1, other RBPs

Translational stall (ISR, mTOR inhibition)

Minutes; mRNA triage and protection in Stress Granules

Alternative Splicing

SR proteins, hnRNPs

Hypoxia, cell signaling, heat shock

Minutes to hours; Proteome diversification, production of stress-specific isoforms

mRNA Stability Control

RNA-Binding Proteins (RBPs), 3' UTR elements, RNA surveillance machinery

Oxidative stress, specific signaling cues

Minutes; Rapid fine-tuning of specific transcript levels, degradation of damaged RNAs

 

The Integrated Stress Response (ISR): A Central Hub for Translational Control

The Integrated Stress Response (ISR) is a pivotal signaling pathway that acts as a central command center for translational control during stress.  It is activated by a remarkably diverse set of stressors, which are detected by four distinct eIF2α kinases. Each kinase possesses unique regulatory domains that allow it to sense a specific type of cellular imbalance, yet they all converge on a single downstream event: the phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eIF2α) on serine 51.

Kinase

Primary Activating Stress Signal(s)

Cellular Location

Downstream Consequence Beyond eIF2α Phosphorylation

PERK

ER stress (unfolded proteins), glucose deprivation, redox imbalance

ER Membrane

Activates the full Unfolded Protein Response (UPR) signaling cascade

GCN2

Amino acid starvation (sensed via uncharged tRNAs), UV light

Cytosol

Directly links nutrient availability to translational control

HRI

Heme deficiency, oxidative stress, heat shock, proteasome inhibition

Cytosol

Couples globin synthesis to heme availability in erythroid cells

PKR

Viral double-stranded RNA (dsRNA), other cellular stresses

Cytosol

Key sensor in the innate antiviral immune response

The phosphorylation of eIF2α has a profound, bifurcated effect on protein synthesis. It converts eIF2 into a potent inhibitor of its own recycling factor, eIF2B, thereby drastically reducing the global rate of translation initiation. This rapid shutdown conserves cellular energy and prevents the synthesis of new proteins under potentially error-prone conditions.  Simultaneously, this global attenuation enables the selective translation of a small subset of mRNAs, most notably that of the transcription factor ATF4. These specialized mRNAs contain regulatory elements called upstream open reading frames (uORFs) in their 5' leaders, which allow them to be efficiently translated precisely when the machinery is stalled, thus activating the downstream transcriptional arm of the stress response. Stress Granules (SGs): Dynamic Hubs for mRNA Triage

A direct consequence of the widespread translational arrest initiated by the ISR is the formation of Stress Granules (SGs). These are dense, non-membranous cytoplasmic bodies formed by the liquid-liquid phase separation of stalled translation pre-initiation complexes, untranslated mRNAs, and a host of RNA-binding proteins (RBPs).  SGs are not static aggregates but highly dynamic compartments that serve as centers for mRNA triage. They function to sequester and protect the bulk of the cell's "housekeeping" mRNAs from degradation during the stress period. Upon resolution of the stress, SGs rapidly dissolve, releasing the stored mRNAs to re-engage with ribosomes, which facilitates a swift return to normal protein synthesis and cellular function.  In addition to this storage function, SGs also act as signaling hubs, concentrating signaling molecules and enzymes, such as the kinase PKR, to amplify the stress response. Alternative Splicing (AS) Under Duress

Alternative splicing is a fundamental post-transcriptional process that dramatically expands the coding capacity of the genome. By differentially including or excluding exons from a pre-mRNA transcript, a single gene can give rise to multiple distinct protein isoforms, often with different functions, localizations, or stabilities.  This process is not static but is dynamically regulated in response to cellular signals and environmental cues. Metabolic stressors, particularly hypoxia, are potent modulators of alternative splicing.  Studies have shown that hypoxia can trigger thousands of alternative splicing events, profoundly altering the cellular proteome.  These stress-induced splicing changes are mediated by alterations in the expression, localization, or activity of core splicing regulators, such as the Serine/Arginine-rich (SR) proteins and heterogeneous nuclear ribonucleoproteins (hnRNPs). The consequences of stress-induced AS can be adaptive or pathological. For example, splicing changes can generate protein isoforms that promote cell survival under hypoxic conditions. However, chronic or aberrant splicing is a hallmark of disease. In neurodegeneration, hypoxia-induced changes in the splicing of APP and tau pre-mRNAs can promote the production of the toxic protein variants that drive Alzheimer's disease pathology.  In oncology, hypoxia-induced alternative splicing is now recognized as a major driver of tumor progression, angiogenesis, and metastasis, and the splicing machinery itself is being explored as a therapeutic target. mRNA Stability and Turnover: Regulating Transcript Half-Life

The abundance of a protein is determined not only by its rate of synthesis but also by the degradation rate of its corresponding mRNA. The regulation of mRNA stability, or half-life, is therefore a critical layer of post-transcriptional control that allows for rapid adjustments in gene expression.  The stability of an mRNA is often dictated by specific sequence elements within its 3' UTR, which serve as binding sites for various RBPs that can either protect the transcript from degradation (e.g., HuR) or target it to decay pathways (e.g., AUF1). Oxidative stress has a particularly profound impact on mRNA stability. RNA molecules are highly susceptible to damage by ROS, with the hydroxyl radical (OH•) being a major culprit.  The most common form of oxidative RNA damage is the conversion of guanosine to 8-hydroxyguanosine (8-OHG).  The presence of such lesions on an mRNA can cause ribosomes to stall during translation, leading to reduced protein output.  To prevent the accumulation of damaged transcripts and the potential production of aberrant proteins, cells have evolved sophisticated RNA surveillance pathways. These systems, analogous to nonsense-mediated decay (NMD), recognize and rapidly degrade oxidized mRNAs.  In some cases, specific RNA structures, such as RNA G-quadruplexes (rG4s), can function as direct ROS sensors; oxidative damage to the structure leads to its unfolding and subsequent destabilization of the entire mRNA molecule.

The Epitranscriptome: A New Frontier in Stress Regulation

Beyond the regulation of transcription and processing, a further layer of control exists directly on the RNA molecules themselves: the epitranscriptome. This refers to the collective of over 140 distinct, enzymatically catalyzed chemical modifications that adorn RNA, creating a dynamic regulatory code that influences every step of an RNA's life cycle. This field has revealed that RNA is not just a passive carrier of information but is actively decorated with marks that are written, erased, and read to control gene expression in response to cellular cues.

An Introduction to RNA Modifications: The "Writers, Readers, and Erasers"

     The epitranscriptome is managed by a dedicated set of proteins that function in a manner analogous to the epigenetic machinery that modifies chromatin. Writers are enzymes, such as methyltransferases, that install a specific chemical mark onto an RNA nucleotide. A prime example is the METTL3-METTL14 complex, which catalyzes the m6A modification.

     Erasers are enzymes that remove these marks, allowing the process to be reversible and dynamic. The FTO and ALKBH5 proteins, for instance, are demethylases that remove the m6A mark.

     Readers are proteins that contain specialized domains to specifically recognize and bind to a modified nucleotide. This binding event is what translates the chemical mark into a biological outcome, such as altering the RNA's stability, localization, splicing, or translation. The YTH-domain family of proteins are well-characterized readers of m6A.

The dysregulation of this machinery is increasingly linked to a wide array of human diseases, including cancer, neurological disorders, and metabolic diseases. N6-methyladenosine (m6A): The Preeminent mRNA Mark

Among the many RNA modifications, N6-methyladenosine (m6A) is the most abundant and well-studied internal modification on eukaryotic mRNA.  It is typically deposited within a consensus sequence motif (RRACH, where R=A/G, H=A/C/U) and is enriched in specific transcript regions, such as near stop codons and within long internal exons and 3' UTRs. The m6A pathway is a critical regulator of the cellular response to metabolic stress.  Its role is particularly prominent in the control of lipid metabolism and associated pathologies like obesity and non-alcoholic fatty liver disease (NAFLD).  The gene encoding the m6A eraser FTO was famously the first to be identified as a major obesity-risk locus through genome-wide association studies. Mechanistically, FTO can promote adipogenesis and lipid accumulation by removing m6A marks from the mRNAs of key lipogenic transcription factors, such as

PPARγ and SREBP1c. This demethylation increases the stability of these transcripts, leading to higher protein levels and enhanced fat storage.  The functional outcome of an m6A mark is context-dependent and determined by the specific reader protein that binds it. For example, binding by the reader YTHDF2 often targets the m6A-modified mRNA for accelerated degradation, whereas binding by YTHDF1 can enhance its translation. A particularly elegant principle of cellular regulation is revealed by the co-regulation of the epigenome and the epitranscriptome. The enzymes that control these two systems are often dependent on the same pool of intermediary metabolites. For instance, the m6A writer METTL3 uses SAM as its methyl donor, the same metabolite used by histone and DNA methyltransferases. Similarly, the m6A erasers FTO and ALKBH5 are αKG-dependent dioxygenases, just like the TET and JmjC demethylases that act on chromatin. This means that a single metabolic shift, such as a decrease in αKG levels during hypoxia, can simultaneously inhibit demethylation of both histones and RNA. This creates a powerful, coordinated, multi-layered response that ensures the cell's entire gene expression program, from transcription to RNA fate, is coherently modulated by its metabolic state. This deep integration underscores that these are not separate systems but rather two arms of a single, integrated network responding to the same upstream metabolic signals.

Other Key Modifications and RNA Editing

While m6A is the most studied mRNA modification, other epitranscriptomic marks and processes are also vital for the stress response.

     Adenosine-to-Inosine (A-to-I) Editing: This process, catalyzed by the ADAR family of enzymes, converts adenosine to inosine within regions of double-stranded RNA.  Since the cellular machinery interprets inosine as guanosine, this modification can have profound effects, including altering protein-coding sequences, creating or removing splice sites, and changing the targeting specificity of miRNAs.  A-to-I editing is essential for normal neurological function and has been implicated in the response to oxidative stress.

     Pseudouridylation (Ψ): This is the enzymatic isomerization of uridine into a different isomer, pseudouridine. This modification can alter the structure of RNA and has been shown to enhance the stability and translational efficiency of mRNA.  The strategic incorporation of pseudouridine into synthetic mRNAs was a breakthrough that enabled the development of the highly stable and effective mRNA vaccines for COVID-19.

It is critical to distinguish these precise, enzyme-catalyzed regulatory events from the stochastic damage that RNA sustains from uncontrolled oxidative stress. Programmed RNA editing and modification are sophisticated mechanisms of gene regulation, whereas oxidative RNA damage is a pathological event that the cell must either repair or eliminate.

Non-Coding RNAs as Master Regulators of Metabolic Homeostasis

Within the vast non-coding transcriptome, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) stand out as particularly influential regulators of the metabolic stress response. They form complex networks that interpret stress signals and execute specific gene expression programs, acting as both fine-tuners and master switches of cellular fate. A functional duality appears to exist between these two classes: miRNAs generally act as rheostats, providing subtle, network-wide adjustments to maintain stability, while lncRNAs often function as architects, orchestrating major, decisive shifts in cell state.

MicroRNAs (miRNAs): Fine-Tuning Metabolic Networks

miRNAs are small non-coding RNAs that function as post-transcriptional repressors, typically by binding to the 3' UTR of target mRNAs to trigger their degradation or inhibit their translation.  A single miRNA can regulate hundreds of different target genes, allowing them to act as powerful rheostats that fine-tune the output of entire metabolic pathways and signaling networks.  Their expression is highly dynamic and responsive to metabolic cues, and their dysregulation is a common feature of metabolic diseases. Hypoxia is a particularly potent regulator of miRNA expression and function.  It induces a characteristic set of miRNAs, collectively termed "hypoxamirs," which play a central role in cellular adaptation to low oxygen. The canonical hypoxamir is

miR-210, which is directly transcribed in response to HIF-1α activation. miR-210 orchestrates the metabolic switch toward glycolysis by targeting and repressing key components of the mitochondrial TCA cycle and electron transport chain, thereby suppressing oxygen consumption and promoting cell survival under hypoxic conditions. Beyond hypoxia, miRNAs are critical regulators of glucose and lipid homeostasis. For example:

     miR-33a and miR-33b are located within the introns of the genes encoding the SREBP transcription factors, the master regulators of cholesterol and fatty acid synthesis. The miRNAs are co-transcribed with their host genes and act in concert with them to control lipid metabolism, creating a coherent feed-forward regulatory loop.

     miR-375 is highly expressed in pancreatic islets and is a key regulator of insulin secretion and the maintenance of β-cell mass. Its dysregulation is strongly implicated in the pathogenesis of diabetes.

     miR-34a is induced in pancreatic β-cells by lipotoxic stress (exposure to high levels of saturated fatty acids) and promotes apoptosis by repressing the anti-apoptotic protein Bcl2, thus contributing to β-cell loss in T2DM.

Because they are stable molecules that can be detected in circulating biofluids like blood plasma, miRNAs are also emerging as highly promising non-invasive biomarkers for monitoring metabolic stress and disease progression. Long Non-Coding RNAs (lncRNAs): Versatile Orchestrators of the Stress Response

lncRNAs are a large and functionally diverse class of transcripts that have emerged as major players in the regulation of cellular stress responses, including metabolic, oxidative, and genotoxic stress.  They achieve their regulatory effects through a wide array of molecular mechanisms. In the nucleus, many lncRNAs function as guides or scaffolds, recruiting chromatin-modifying complexes like Polycomb Repressive Complex 2 (PRC2) to specific gene loci to epigenetically silence their expression.  In the cytoplasm, they can act as "sponges" or decoys for miRNAs, binding and sequestering them to prevent them from repressing their own mRNA targets. Numerous lncRNAs have been specifically implicated in metabolic stress and disease:

     MALAT1 is upregulated in the context of diabetic complications and has been shown to promote hepatic steatosis (fatty liver) and insulin resistance. It also contributes to hyperglycemia-induced damage in retinal cells, linking it to diabetic retinopathy.

     GAS5 (Growth Arrest-Specific 5) often functions as a tumor suppressor and is involved in the cellular response to nutrient deprivation by modulating key pathways like autophagy and apoptosis.

     H19 is an imprinted lncRNA that plays a complex role in hepatic glucose metabolism; its inhibition has been shown to impair insulin signaling and increase the expression of gluconeogenic genes.

Stress-responsive lncRNAs are now understood to be critical mediators of the pathological effects of hyperglycemia-induced oxidative and ER stress, which are the primary drivers of diabetic complications such as nephropathy (kidney disease) and retinopathy (eye disease).  This regulatory duality—miRNAs as fine-tuners and lncRNAs as master switches—suggests a sophisticated division of labor in the cell's regulatory architecture, with different therapeutic implications. Re-balancing a dysregulated pathway might be achieved by targeting a miRNA, whereas reversing a pathologically entrenched cell state might require intervention at the level of a key lncRNA.

Integration and Pathophysiological Implications

The intricate RNA-based regulatory mechanisms described—from transcriptional reprogramming to post-transcriptional control and epitranscriptomic marking—do not operate in isolation. They are woven into a cohesive network that is coordinated by master signaling hubs, which integrate information about the cell's metabolic state and direct the appropriate response. The chronic dysregulation of this integrated network is a fundamental driver of pathology in a host of human diseases. Understanding this system-level behavior is key to developing effective therapeutic strategies.

Signaling Hubs: The AMPK/mTOR Axis

     At the heart of cellular metabolic regulation lie two evolutionarily conserved protein kinase signaling pathways: the AMP-activated protein kinase (AMPK) pathway and the mechanistic target of rapamycin (mTOR) pathway. These two hubs act as a central processing unit, sensing the cell's energy and nutrient status and making critical decisions about cellular growth and metabolism. They function as counter-regulators: AMPK is the guardian of catabolism, while mTOR is the governor of anabolism. AMPK (The Energy Sensor): AMPK is activated by conditions of low cellular energy, signified by a high ratio of AMP to ATP, such as during exercise or nutrient deprivation.  Once active, AMPK phosphorylates a multitude of substrates to switch on catabolic, ATP-producing pathways (like fatty acid oxidation and autophagy) and switch off anabolic, ATP-consuming processes (like protein and lipid synthesis).

     mTOR (The Nutrient Sensor): The mTOR kinase is the core component of two distinct protein complexes, mTORC1 and mTORC2. mTORC1 is the primary nutrient-sensing complex, activated by high levels of nutrients (especially amino acids) and growth factors.  Active mTORC1 drives cell growth and proliferation by promoting anabolic pathways and potently inhibiting catabolic processes like autophagy.

Crucially, the influence of AMPK and mTOR extends far beyond the direct phosphorylation of metabolic enzymes. These kinases are deeply intertwined with the RNA regulatory machinery.  Both kinases can translocate to the nucleus, where they directly phosphorylate transcription factors and chromatin-modifying enzymes to control gene expression programs.  They also exert powerful control over the translation machinery through phosphorylation of key targets like S6 kinase (S6K) and 4E-binding protein 1 (4E-BP1), which in turn dictates global protein synthesis rates and the formation of stress granules.  The interplay between these two pathways is managed by extensive reciprocal inhibition: AMPK directly phosphorylates and inhibits mTORC1, while mTORC1 can phosphorylate and suppress AMPK, creating a sensitive molecular switch that fine-tunes the cell's metabolic posture. Dysregulation in Disease: When Adaptation Becomes Pathology

     While the activation of these RNA-centric stress responses is essential for surviving acute challenges, their chronic, unabated activation in the face of persistent metabolic stress is a primary driver of disease. Metabolic Syndrome and T2DM: The state of chronic hyperglycemia and hyperlipidemia (glucolipotoxicity) that characterizes T2DM and metabolic syndrome creates a vicious cycle of persistent ER stress and oxidative stress. This leads to the pathological dysregulation of numerous ncRNAs. For instance, specific miRNAs and lncRNAs are induced that promote pancreatic β-cell apoptosis, drive insulin resistance in peripheral tissues, and fuel chronic low-grade inflammation.  Furthermore, dysregulation of the m6A epitranscriptomic machinery, particularly involving the obesity-associated geneFTO, is a key contributor to aberrant lipid metabolism and insulin resistance.

     Neurodegenerative Diseases: The proper regulation of RNA processing is paramount for the health and function of long-lived, post-mitotic cells like neurons, and its dysregulation is a common theme in neurodegenerative diseases such as AD, Parkinson's disease, and ALS.  Metabolic stress is a major risk factor for these conditions, contributing to pathology through multiple RNA-based mechanisms, including aberrant alternative splicing of disease-relevant genes liketau and APP  , direct oxidative damage to RNA molecules  , and the dysregulation of epitranscriptomic marks that are essential for synaptic function and neuronal survival.

     Cancer: Cancer cells must survive and proliferate within the harsh and metabolically stressful tumor microenvironment, which is often characterized by hypoxia and nutrient limitation. To do so, they hijack and co-opt the cell's innate stress response pathways. They undergo profound metabolic reprogramming (e.g., the Warburg effect) and become dependent on stress-activated ncRNAs and other RNA regulators to evade apoptosis, sustain proliferation, and enable invasion and metastasis.

The table below highlights select non-coding RNAs whose dysregulation in specific metabolic stress contexts contributes to disease.

 

ncRNA Name

Class

Metabolic Stress Context

Key Target(s)/Mechanism

Contribution to Pathology

miR-34a

miRNA

Lipotoxicity (high fatty acids)

Targets anti-apoptotic Bcl2 and insulin secretion factor VAMP2

Promotes pancreatic β-cell apoptosis in T2DM

miR-210

miRNA

Hypoxia

Induced by HIF-1α; targets mitochondrial OXPHOS components

Drives glycolytic switch in tumors, promoting survival and angiogenesis

MALAT1

lncRNA

Hyperglycemia, Lipotoxicity

Promotes hepatic steatosis; may sponge miRNAs

Contributes to diabetic retinopathy and insulin resistance

GAS5

lncRNA

Nutrient deprivation

Modulates autophagy and apoptosis via mTOR signaling pathway

Acts as a tumor suppressor; its loss contributes to cancer progression

 

Therapeutic Horizons: Targeting RNA Pathways

     The central and causal role of RNA dysregulation in metabolic disease makes these pathways highly attractive for therapeutic intervention.  A new generation of RNA-based medicines is emerging that can directly target RNA molecules with high specificity, moving beyond traditional small molecule and antibody drugs. Oligonucleotide Therapeutics: Synthetic strands of nucleic acids, such as small interfering RNAs (siRNAs) and antisense oligonucleotides (ASOs), can be designed to bind to a specific mRNA or lncRNA via complementary base pairing. This binding can trigger the transcript's degradation or modulate its splicing, effectively silencing a disease-causing gene. The development of chemical modifications and delivery technologies, such as conjugation to N-acetylgalactosamine (GalNAc) for liver-specific uptake, has led to approved therapies for several genetic liver metabolic diseases.

     mRNA Therapy: As famously demonstrated by the COVID-19 vaccines, lipid nanoparticles can be used to deliver synthetic mRNA into cells, instructing them to produce a specific protein. This approach holds great promise for protein replacement therapies in rare genetic diseases and offers a potentially safer alternative to viral vector-based gene therapy due to a reduced risk of genomic integration.

     Targeting ncRNAs: Strategies are being developed to therapeutically modulate ncRNAs. "Antagomirs" are modified oligonucleotides that can bind to and inhibit an overactive miRNA, while synthetic miRNA "mimics" can be delivered to restore the function of a miRNA that is lost in a disease state.

Despite this promise, significant challenges remain, including the development of efficient methods for delivering these drugs to tissues beyond the liver, ensuring their long-term safety and efficacy, and addressing the high manufacturing costs associated with these advanced therapies. The profound interconnectedness of the pathways involved in the metabolic stress response suggests that the most powerful therapeutic strategies may be those that target not a single downstream effector molecule, but a critical regulatory node that controls the entire network's state. The AMPK/mTOR signaling axis is a prime example of such a node. Modulating the activity of AMPK or mTOR with a small molecule drug has the potential to coordinately reset multiple downstream arms of the RNA-based response—rebalancing miRNA profiles, altering lncRNA expression, and shifting translational programs—in a holistic manner. This shifts the therapeutic paradigm from a "one gene, one drug" approach to a "one network, one drug" philosophy, highlighting the future importance of systems biology in developing treatments for complex, multifactorial diseases.

Conclusion

The cellular response to metabolic stress is a masterclass in regulatory complexity, orchestrated in large part by the diverse and dynamic world of RNA. Far from being a simple linear pathway, the response is a multi-layered, interconnected network where RNA acts as a central processor, sensing core cellular perturbations and executing a sophisticated, temporally organized defense. Transcriptional reprogramming, driven by stress-responsive transcription factors and a chromatin landscape that directly senses the cell's metabolic state, establishes a new, long-term cellular identity. This foundational change is preceded and supported by a suite of rapid post-transcriptional mechanisms—including translational control via the Integrated Stress Response, mRNA triage in stress granules, and dynamic changes in alternative splicing and stability—that allow the cell to weather acute insults.

Layered on top of this is the epitranscriptome, a dynamic chemical code written onto RNA itself, which is co-regulated with the epigenome by the same intermediary metabolites, ensuring a coherent, system-wide response. This entire network is fine-tuned and directed by non-coding RNAs, with miRNAs acting as rheostats to buffer pathways and lncRNAs acting as architects to execute major shifts in cell fate. The integration of these RNA-centric pathways by master signaling hubs like the AMPK/mTOR axis underscores the holistic nature of the cellular response.

While these adaptive responses are essential for survival, their chronic dysregulation is a fundamental driver of modern human diseases, from T2DM and neurodegeneration to cancer. The elucidation of these RNA-based mechanisms has not only transformed our understanding of disease pathogenesis but has also opened up exciting new therapeutic frontiers. The development of RNA-based medicines and strategies to target the central nodes of these regulatory networks holds immense promise for treating these complex and debilitating conditions. The future of medicine will undoubtedly involve a deeper appreciation for, and a more precise manipulation of, the RNA-centric nexus of metabolic stress.

#Hashtags

#RNA #MetabolicStress #GeneRegulation #CellularStress #Transcriptomics #Epitranscriptome #m6A #ncRNA #miRNA #lncRNA #Hypoxia #OxidativeStress #NutrientSensing #IntegratedStressResponse #AlternativeSplicing #mRNAStability #AMPK #mTOR #Diabetes #Neurodegeneration #CancerMetabolism #RNATherapeutics #CellBiology #MolecularBiology

References

1.    Role of metabolic stress for enhancing muscle adaptations: Practical ..., https://pmc.ncbi.nlm.nih.gov/articles/PMC5489423/

2.    What Is Metabolic Stress? - Proathlix, https://proathlix.com/blogs/knowledge/what-is-metabolic-stress

3.    Cellular metabolic stress: Considering how cells respond to nutrient excess - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC3190402/

4.    Endocytosis in the adaptation to cellular stress - Cell Stress, http://www.cell-stress.com/researcharticles/2020a-lopez-hernandez-cell-stress/

5.    Brain metabolic stress and neuroinflammation at the basis of cognitive impairment in Alzheimer's disease - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC4436878/

6.    RNA Regulation of Lipotoxicity and Metabolic Stress - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC4915576/

7.    Nutrient Sensing and the Oxidative Stress Response - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC5438757/

8.    ROS signaling under metabolic stress: cross-talk between AMPK and AKT pathway, https://d-nb.info/1130955117/34

9.    Hypoxia-induced alternative splicing: the 11th Hallmark of Cancer - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC7294618/

10.  Short-Term Hypoxia in Cells Induces Expression of Genes Which Are Enhanced in Stressed Cells - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC9498350/

11.  Hypoxia-induced alternative splicing in human diseases: the pledge, the turn, and the prestige - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC11072330/

12.  Oxidative damage to RNA: mechanisms, consequences, and ..., https://pmc.ncbi.nlm.nih.gov/articles/PMC3010397/

13.  The role of oxidative stress in the dysregulation of gene expression and protein metabolism in neurodegenerative disease - PubMed, https://pubmed.ncbi.nlm.nih.gov/16487048/

14.  What are the functions of rRNA, tRNA, mRNA? - Quora, https://www.quora.com/What-are-the-functions-of-rRNA-tRNA-mRNA

15.  Biochemistry, RNA Structure - StatPearls - NCBI Bookshelf, https://www.ncbi.nlm.nih.gov/books/NBK558999/

16.  Ribonucleic Acid (RNA) Fact Sheet, https://www.genome.gov/about-genomics/educational-resources/fact-sheets/ribonucleic-acid-fact-sheet

17.  Post-transcriptional regulation in metabolic diseases - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC3495744/

18.  Post-transcriptional regulation during stress - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC9246287/

19.  Coding and Non-Coding RNA - Bio-Rad, https://www.bio-rad.com/en-us/applications-technologies/coding-non-coding-rna?ID=Q1070M70KWE7

20.  RNA - Wikipedia, https://en.wikipedia.org/wiki/RNA

21.  Types of RNA (article) | Khan Academy, https://www.khanacademy.org/test-prep/mcat/biomolecules/x04f6bc56:transcription-and-translation/a/types-of-rna

22.  Different types of RNAs and their functions - FutureLearn, https://www.futurelearn.com/info/courses/translational-research/0/steps/14201

23.  Noncoding RNA molecules | EBSCO Research Starters, https://www.ebsco.com/research-starters/health-and-medicine/noncoding-rna-molecules

24.  Roles of RNA in Biology - RNA Therapeutics Institute - UMass Chan Medical School, https://www.umassmed.edu/rti/biology/what-is-rna/role-of-rna-in-biology/

25.  Non-coding RNA - Wikipedia, https://en.wikipedia.org/wiki/Non-coding_RNA

26.  MicroRNAs in Metabolism and Metabolic Disorders - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC4021399/

27.  New regulation mechanisms of natural products and microRNA on metabolic syndrome, https://www.spkx.net.cn/EN/abstract/abstract39422.shtml

28.  Long Noncoding RNAs in Metabolic Syndrome Related Disorders - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC5110871/

29.  Long non-coding RNAs: roles in cellular stress responses and epigenetic mechanisms regulating chromatin, https://pmc.ncbi.nlm.nih.gov/articles/PMC11123517/

30.  Full article: Long non-coding RNAs: roles in cellular stress responses and epigenetic mechanisms regulating chromatin, https://www.tandfonline.com/doi/full/10.1080/19491034.2024.2350180

31.  Long Non-coding RNAs: Major Regulators of Cell Stress in Cancer - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC7099402/

32.  Long Non-coding RNAs: Major Regulators of Cell Stress in Cancer - PubMed, https://pubmed.ncbi.nlm.nih.gov/32266130/

33.  Hypoxia: A master regulator of microRNA biogenesis and activity ..., https://msesiom2.demo.elsevierpure.com/en/publications/hypoxia-a-master-regulator-of-microrna-biogenesis-and-activity

34.  Mastering Transcriptional Regulation, https://www.numberanalytics.com/blog/mastering-transcriptional-regulation-molecular-cell-signaling

35.  New insights into transcriptional reprogramming during cellular ..., https://journals.biologists.com/jcs/article/132/21/jcs238402/224639/New-insights-into-transcriptional-reprogramming

36.  Hypoxia-Induced Alternative Splicing in Endothelial Cells | PLOS One, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0042697

37.  The Regulation of Integrated Stress Response Signaling Pathway on Viral Infection and Viral Antagonism - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC8874136/

38.  The integrated stress response | EMBO reports - EMBO Press, https://www.embopress.org/doi/10.15252/embr.201642195

39.  Metabolic regulation of chromatin modifications and gene ..., https://rupress.org/jcb/article/217/7/2247/39126/Metabolic-regulation-of-chromatin-modifications

40.  Stress and its impact on the transcriptome - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC8213869/

41.  Circulating cell-free RNA signatures for the characterization and diagnosis of myalgic encephalomyelitis/chronic fatigue syndrome | PNAS, https://www.pnas.org/doi/10.1073/pnas.2507345122

42.  The Stress Granule Protein G3BP1 Recruits Protein Kinase R To Promote Multiple Innate Immune Antiviral Responses - ASM Journals, https://journals.asm.org/doi/10.1128/jvi.02791-14

43.  non-stop decay mRNA surveillance pathway is required for oxidative stress tolerance | Nucleic Acids Research | Oxford Academic, https://academic.oup.com/nar/article/45/11/6881/3787808

44.  The Chemistry Behind mRNA Stability - Number Analytics, https://www.numberanalytics.com/blog/chemistry-behind-mrna-stability-nucleic-acids

45.  RNA G-Quadruplex within the 5'-UTR of FEN1 Regulates mRNA Stability under Oxidative Stress - PubMed, https://pubmed.ncbi.nlm.nih.gov/36829835/

46.  The epitranscriptome in ageing and stress resistance: A systematic review - PubMed, https://pubmed.ncbi.nlm.nih.gov/35908668/

47.  Dawn of Epitranscriptomic Medicine | Circulation - American Heart Association Journals, https://www.ahajournals.org/doi/10.1161/CIRCGEN.118.001927

48.  Regulatory role and mechanism of m6A RNA modification in human metabolic diseases, https://pmc.ncbi.nlm.nih.gov/articles/PMC8399361/

49.  Epitranscriptomics in the Heart: A Focus on m6A - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC7486268/

50.  Context-Dependent Roles of RNA Modifications in Stress Responses and Diseases - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC7920320/

51.  Decoding RNA Metabolism in Disease - Number Analytics, https://www.numberanalytics.com/blog/rna-metabolism-disease-transcriptomics

52.  Metabolic Stress and Somatic Mutation: Exploring Links to Sleep Disorders, https://ntrs.nasa.gov/citations/20250002640

53.  New developments in AMPK and mTORC1 cross-talk | Essays in Biochemistry, https://portlandpress.com/essaysbiochem/article/68/3/321/234702/New-developments-in-AMPK-and-mTORC1-cross-talk

54.  Insight on Transcriptional Regulation of the Energy Sensing AMPK and Biosynthetic mTOR Pathway Genes - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC7438746/

55.  RNA dysregulation in neurodegenerative diseases - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC11790913/

56.  Deregulated transcription factors in cancer cell metabolisms and reprogramming - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC11220368/

57.  RNA-based therapies in liver metabolic diseases - PubMed, https://pubmed.ncbi.nlm.nih.gov/39988358/

Featured Story

How RNA Regulates Metabolic Stress