Fact-checked by Grok 2 weeks ago

Cellular stress response

The cellular stress response encompasses the suite of adaptive molecular mechanisms that cells activate to detect, counteract, and recover from insults that disrupt homeostasis, including environmental stressors like heat, oxidative damage, hypoxia, and genotoxic agents, as well as intrinsic challenges such as protein misfolding or metabolic imbalances. These responses aim to repair damaged macromolecules—such as proteins, DNA, RNA, and lipids—or, if damage is irreparable, to initiate programmed cell death pathways like apoptosis to prevent propagation of harm. Evolutionarily conserved across all organisms, this response forms a universal defense system, often termed the "minimal stress proteome," which integrates sensing, signaling, and effector functions to balance survival and elimination. Central to these mechanisms are specialized pathways tailored to specific stressors. The heat shock response (HSR), triggered by elevated temperatures or protein denaturation, induces transcription factors like HSF1 to upregulate heat shock proteins (HSPs), such as and , which act as molecular chaperones to refold misfolded proteins and inhibit aggregation. Similarly, the unfolded protein response (UPR) addresses (ER) stress from accumulated unfolded proteins via three main sensors—IRE1, PERK, and ATF6—that enhance capacity, reduce translation to alleviate ER load, or activate antioxidants like Nrf2 to combat . The DNA damage response (DDR) employs kinases such as and ATR to halt the , recruit repair enzymes, or activate p53-mediated if lesions persist, safeguarding genomic integrity. Other pathways, including those for (e.g., Nrf2-mediated antioxidant induction) and (e.g., HIF signaling), converge on common effectors like for degrading damaged components or regulation for metabolic reprogramming. These responses are pivotal for physiological and prevention but can become dysregulated in . In healthy contexts, they maintain tissue , support , and enhance to transient stresses, such as during exercise or . However, chronic activation or impairment contributes to conditions like cancer, where upregulated HSR and UPR promote tumor survival and therapy resistance; neurodegenerative disorders, such as Parkinson's, where failed protein clearance leads to aggregation; and cardiovascular diseases, including , via unresolved oxidative damage. Aging further diminishes response efficiency, exacerbating vulnerability to stress. Overall, the cellular stress response exemplifies a delicate "balancing act," deciding between repair and based on stress intensity and duration to optimize organismal fitness.

Overview

Definition and importance

The cellular stress response refers to a conserved adaptive mechanism by which cells detect and counteract damaging conditions, such as protein misfolding, oxidative damage, or energy depletion, to restore and promote survival. This response encompasses a range of molecular pathways that enable cells to sense macromolecular perturbations and initiate protective measures, including the upregulation of chaperones and repair systems, while conserving resources during adversity. The importance of the cellular stress response lies in its critical role in preventing , enhancing cellular resilience to environmental insults, and maintaining —the dynamic regulation of , trafficking, and . By inhibiting pro-apoptotic processes like activation and release, these mechanisms allow cells to survive transient es that might otherwise lead to . Furthermore, the response is evolutionarily conserved across prokaryotes and eukaryotes, with a core set of approximately 44 proteins forming a minimal proteome that underscores its fundamental necessity for life, from to humans. This phenomenon was first observed in 1962 by Ferruccio Ritossa, who noted heat-induced chromosomal puffs in Drosophila melanogaster salivary glands, revealing a novel pattern of gene activity that led to the identification of stress-inducible genes and the broader field of stress responses. Overall, cellular stress responses integrate multiple signaling pathways to finely balance survival strategies against the risk of irreparable damage, ensuring organismal health and adaptation.

Types of cellular stress

Cellular stress can be broadly classified into physical, chemical, metabolic, and endogenous categories, each representing distinct inputs that challenge cellular homeostasis and trigger adaptive responses. Physical stressors involve external forces such as elevated temperatures or ionizing radiation, which directly disrupt molecular structures. Chemical stressors encompass exogenous toxins and heavy metals that interfere with biochemical processes. Metabolic stressors arise from imbalances in energy availability or oxygen supply, while endogenous stressors originate from internal cellular dysfunctions like protein misfolding or genetic insults. This classification highlights the diversity of threats cells face, from environmental exposures to intrinsic errors, and underscores the need for tailored protective mechanisms. Physical stressors, including and , primarily affect protein stability and genomic integrity. Heat stress occurs when temperatures rise 3–5°C above physiological norms, leading to protein denaturation where unfolded polypeptides aggregate and impair cellular function. For instance, exposure to temperatures exceeding 42°C rapidly destabilizes proteins, initiating protective pathways to refold or degrade damaged molecules. , such as UV or ionizing types, induces physical breaks in DNA strands, compromising replication and transcription. These stressors often manifest acutely, like sudden heat exposure, overwhelming cellular thresholds if not resolved promptly. Chemical stressors involve xenobiotics and heavy metals that bind cellular components or generate toxic byproducts. Toxins like pesticides or industrial pollutants disrupt enzymatic activities and membrane integrity, while heavy metals such as arsenic accumulate and catalyze harmful reactions. Arsenic, a common environmental xenobiotic, binds to sulfhydryl groups in proteins, but cells counter this by inducing metallothioneins—cysteine-rich proteins that sequester the metal and mitigate toxicity. Oxidative stress, a frequent outcome of chemical exposure, stems from an imbalance where reactive oxygen species (ROS) production exceeds antioxidant capacity, damaging lipids, proteins, and DNA. Chronic chemical exposure, unlike acute bursts, leads to cumulative bioaccumulation, lowering cellular resilience over time. Metabolic stressors disrupt energy homeostasis through oxygen or nutrient shortages. Hypoxia, or oxygen deprivation, impairs mitochondrial respiration, forcing cells to shift to less efficient anaerobic pathways and accumulate metabolic byproducts. Nutrient deprivation, such as glucose starvation, halts ATP production and triggers catabolic processes to sustain viability. These often overlap with endogenous origins in pathological states like ischemia. Endogenous stressors emerge from internal perturbations, including endoplasmic reticulum (ER) overload and DNA damage. ER overload occurs when protein synthesis exceeds folding capacity, causing accumulation of misfolded proteins and activating the unfolded protein response. DNA damage, arising from replication errors or repair failures, threatens genomic stability and can propagate if unchecked. Unlike acute physical or chemical insults, chronic endogenous stress, such as persistent ROS imbalance, erodes cellular thresholds progressively, contributing to long-term dysfunction. A key distinction exists between acute and chronic stressors, influencing their impact on cellular thresholds. Acute stressors, like sudden or exposure, elicit rapid, transient responses that restore if mild, but severe cases can exceed repair capacity. stressors, such as ongoing oxidative damage or , impose cumulative effects, gradually depleting reserves and heightening vulnerability to further insults. This temporal aspect determines whether cells adapt or succumb.

Molecular Mechanisms

Protein quality control systems

Protein quality control systems are essential mechanisms that maintain by assisting in the refolding of misfolded proteins and targeting irreparably damaged ones for degradation during cellular stress. These systems primarily involve molecular chaperones and proteolytic pathways, which prevent and ensure cellular . Heat shock proteins (HSPs), such as and , play central roles in this process by recognizing and binding to exposed hydrophobic regions of unfolded or denatured polypeptides, thereby inhibiting aggregation and facilitating refolding. HSP70 operates in an ATP-dependent manner, where it captures substrate proteins in their ATP-bound state with low affinity and, upon ATP hydrolysis stimulated by co-chaperones like HSP40 (also known as DnaJ), transitions to a high-affinity ADP-bound conformation that holds the substrate for refolding or handover to other chaperones. HSP40 co-chaperones initiate this cycle by delivering substrates to HSP70 and accelerating ATP hydrolysis, enabling iterative folding attempts. In contrast, HSP90 primarily stabilizes partially folded proteins and matures client proteins, often in complex with co-chaperones, to prevent aggregation under stress conditions. The simplified chaperone cycle can be represented as: \text{Unfolded protein} + \text{HSP70 (ATP-bound)} \xrightarrow{\text{HSP40, ATP hydrolysis}} \text{Folded protein} + \text{HSP70 (ADP-bound)} + \text{P}_\text{i} This cycle underscores the energy-dependent nature of chaperone-mediated proteostasis. When refolding fails, degradation pathways clear misfolded proteins to avert toxicity. The ubiquitin-proteasome system (UPS) handles targeted degradation of individual soluble proteins, where E3 ubiquitin ligases tag substrates with ubiquitin chains, marking them for breakdown by the 26S proteasome. This process is crucial during stress, as it selectively removes damaged proteins to restore proteostasis. Complementing the UPS, the autophagy-lysosomal pathway mediates bulk clearance of protein aggregates and organelles by engulfing them into autophagosomes that fuse with lysosomes for degradation. Selective autophagy, such as aggrephagy, targets aggregates specifically, ensuring efficient removal under proteotoxic stress. Together, these systems provide a robust defense against protein misfolding, with their expression often upregulated transcriptionally during stress to enhance capacity.

Transcriptional and translational regulation

Cellular stress responses involve intricate mediated by key s that activate protective gene networks. Heat shock factor 1 (HSF1) plays a central role in the by undergoing stress-induced trimerization and nuclear translocation, enabling it to bind heat shock elements (HSEs) in promoter regions of target genes such as those encoding molecular chaperones. This activation is tightly controlled, with HSF1 monomers maintained in an inactive state under normal conditions through interactions with chaperones like HSP90. Similarly, nuclear factor erythroid 2-related factor 2 (NRF2) regulates responses by dissociating from its inhibitor under stress, allowing NRF2 to translocate to the and bind antioxidant response elements (AREs) to induce genes involved in and balance. In endoplasmic reticulum (ER) stress, activating 4 () and X-box binding protein 1 () coordinate the unfolded protein response (UPR); is transcriptionally upregulated to drive genes for amino acid metabolism and control, while , particularly its spliced isoform (XBP1s), promotes ER expansion and chaperone expression. Translational regulation during stress is exemplified by the integrated stress response (ISR), a conserved pathway that attenuates global protein synthesis while selectively enhancing translation of specific mRNAs. The is triggered by of 2 alpha (eIF2α) by kinases such as PERK, GCN2, PKR, or HRI, which inhibits the formation of the ternary complex required for cap-dependent translation initiation, thereby reducing overall translation by up to 80-90%. However, this paradoxically boosts translation of ATF4 mRNA through its upstream open reading frames (uORFs), where delayed reinitiation favors scanning to the main ATF4 coding sequence under high eIF2α levels. This selective mechanism ensures rapid production of , which then transcriptionally activates UPR target genes, linking translational control directly to downstream transcriptional outputs. Feedback mechanisms prevent overactivation of these pathways, maintaining cellular . For instance, newly synthesized heat shock proteins (HSPs), such as , bind and inhibit HSF1 trimerization and DNA-binding activity, forming a loop that attenuates the once proteotoxic stress is resolved. In the , dephosphorylation of eIF2α by phosphatases like PPP1R15A (GADD34), which is itself induced by , restores global translation and terminates the response. Recent studies highlight the ISR's role in chronic stress adaptation via the PERK-eIF2α- axis, where sustained low-level activation promotes resilience against prolonged proteotoxic or metabolic insults without triggering , as seen in models of neurodegeneration and metabolic disorders post-2020. These regulatory layers ensure that transcriptional and translational adjustments are both protective and reversible, with downstream effectors like chaperones reinforcing protein quality control.

Responses to Specific Stressors

Heat shock response

The (HSR) is a conserved cellular defense mechanism activated primarily by elevated temperatures, resulting in the transcriptional upregulation of heat shock proteins (HSPs) to mitigate protein damage and maintain . This response is orchestrated by heat shock factor 1 (HSF1), a that, under normal conditions, exists in an inactive monomeric form bound to HSPs such as and HSP90. Upon thermal stress, misfolded or aggregated proteins accumulate, disrupting these inhibitory complexes and allowing HSF1 to trimerize, translocate to the , and bind to heat shock elements (HSEs) in the promoters of HSP genes, thereby inducing their expression. The HSR threshold in mammalian cells typically activates around 42°C, varying slightly by cell type and species, marking the transition from mild to severe proteotoxic stress. Central to the HSR are molecular chaperones like and , which play critical roles in cytoprotection by refolding denatured proteins, preventing aggregation, and facilitating the degradation of irreparable ones via the ubiquitin-proteasome system. These HSPs also confer anti-apoptotic effects by inhibiting key executioners of , such as activation and Bax/Bak oligomerization at the mitochondria, thereby preserving cell viability during acute heat exposure. For instance, directly sequesters apoptosis-inducing factor (AIF) and inhibits JNK signaling, while stabilizes client proteins involved in survival pathways, collectively enhancing cellular resilience. The protective outcomes of the HSR extend beyond immediate survival, as demonstrated by preconditioning strategies where mild heat stress (e.g., 42–43°C for short durations) induces HSP expression and confers tolerance to subsequent severe insults like ischemia. In myocardial and neuronal tissues, this preconditioning reduces ischemic by bolstering defenses and limiting , independent of HSP induction in some contexts, highlighting the HSR's broader cardioprotective and neuroprotective potential. Recent studies have expanded the understanding of HSF1's role beyond thermal stress, revealing its activation in non-thermal proteotoxic conditions, such as in neurodegeneration. In models of Alzheimer's and Parkinson's diseases, impaired HSF1 function exacerbates proteotoxicity by failing to upregulate HSPs, leading to accelerated neuronal loss, whereas enhancing HSF1 activity ameliorates aggregate clearance and synaptic integrity. This underscores HSF1's therapeutic promise in age-related proteopathies, with post-2020 research emphasizing its dysregulation in exceptional aging as a key factor in vulnerability to proteotoxic stress.

Oxidative and chemical stress responses

The cellular response to oxidative stress is primarily triggered by reactive oxygen species (ROS), which are generated endogenously from sources such as mitochondrial leaks during and NADPH oxidases during immune signaling and vascular function. These ROS, including anions and , can accumulate and disrupt cellular integrity if not neutralized, prompting adaptive mechanisms to restore balance. A central pathway in this response is the -NRF2 system, where acts as a sensor by detecting oxidative insults through oxidation of its residues, particularly cysteines 151, 273, and 288, leading to conformational changes that release NRF2 from ubiquitination and degradation. Stabilized NRF2 translocates to the and binds antioxidant response elements (AREs) to upregulate genes encoding enzymes, such as () for superoxide dismutation and glutathione S-transferase () for detoxifying products. This NRF2-mediated induction enhances cellular resilience against ROS-induced damage, with converting to less harmful and conjugating to xenobiotics and oxidative byproducts. Chemical stress responses complement oxidative defenses by targeting toxins and through detoxification systems. Phase I enzymes, primarily (CYP450) monooxygenases, introduce reactive groups via oxidation to make xenobiotics more water-soluble, while phase II enzymes like UDP-glucuronosyltransferases (UGTs) conjugate these intermediates with for excretion, preventing and . For , metallothioneins (MTs) bind and sequester ions like and mercury, with their expression induced by the metal-responsive transcription factor 1 (MTF1); for instance, exposure activates MTF1 via binding to metal response elements, elevating MT levels to mitigate metal-induced ROS and protein mishandling. Imbalance in redox , where antioxidant capacity is overwhelmed, promotes —a oxidizing polyunsaturated fatty acids in membranes, generating toxic aldehydes like that propagate damage and amplify stress signaling. Chronic from environmental , such as exposure, sustains this imbalance by persistently elevating ROS in and systemic tissues, contributing to and long-term cellular dysfunction as evidenced in recent analyses of urban air quality impacts.

Hypoxic and ER stress responses

The cellular response to , or oxygen deprivation, primarily involves the stabilization and activation of (HIF-1α), a key that orchestrates metabolic and vascular adaptations to low oxygen levels. Under normoxic conditions, HIF-1α is rapidly degraded through at specific residues by oxygen-dependent prolyl domain () enzymes, which serve as cellular oxygen sensors; this modification allows binding to the von Hippel-Lindau (VHL) complex, targeting HIF-1α for proteasomal degradation. During , reduced oxygen availability inhibits PHD activity, leading to HIF-1α accumulation, dimerization with HIF-1β (also known as ARNT), nuclear translocation, and binding to hypoxia response elements in target gene promoters. This pathway enables cells to shift toward and enhance oxygen delivery. A prominent downstream effect of HIF-1α activation is the upregulation of vascular endothelial growth factor (VEGF), which stimulates endothelial cell proliferation and migration to promote angiogenesis, thereby increasing vascular networks in hypoxic tissues. This response is essential for physiological processes like embryonic development and tissue repair but can drive pathological neovascularization in conditions such as tumors, where sustained hypoxia perpetuates a pro-angiogenic environment. HIF-1α also induces glycolytic enzymes like glucose transporter 1 (GLUT1) and lactate dehydrogenase A (LDHA) to support energy production without oxygen-dependent respiration. Endoplasmic reticulum (ER) stress occurs when the protein folding capacity of the is overwhelmed by unfolded or misfolded proteins, often due to nutrient deprivation, calcium imbalance, or genetic mutations, activating the unfolded protein response (UPR) to restore ER . The UPR is coordinated by three ER transmembrane sensors—PERK (protein kinase R-like ER kinase), IRE1 (inositol-requiring enzyme 1), and ATF6 (activating transcription factor 6)—which are sequestered by the chaperone BiP (, also GRP78) under basal conditions. ER stress causes unfolded proteins to bind BiP, releasing the sensors and initiating parallel signaling branches: PERK phosphorylates eIF2α to globally attenuate translation while selectively enhancing translation of , a that upregulates genes for balance and transport; IRE1 autophosphorylates, activating its endonuclease to splice XBP1 mRNA into a potent that induces ER chaperones, foldases, and ER-associated degradation (ERAD) components; and ATF6 traffics to the Golgi for sequential cleavage by site-1 and site-2 proteases, liberating an N-terminal fragment that translocates to the to drive expression of BiP and . These adaptive mechanisms increase capacity and degrade irreparable proteins.00651-3) If ER stress persists unresolved, the UPR transitions from prosurvival to proapoptotic signaling, primarily through upregulation of C/EBP homologous protein (CHOP, also GADD153), a induced by (via PERK) and ATF6 pathways that sensitizes cells to death by downregulating anti-apoptotic , promoting via GADD34, and enhancing DR5 expression to activate extrinsic . This shift prevents prolonged proteotoxic burden and is implicated in diseases like and neurodegeneration. The integrated response (ISR), which converges on eIF2α phosphorylation through PERK among other kinases, overlaps with UPR to fine-tune under combined hypoxic and ER stresses. Recent 2025 studies reveal that assemblysomes—dynamic RNA-protein complexes—facilitate formation during ISR-UPR crosstalk, sequestering mRNAs for selective translation and protecting cells from acute proteotoxic overload.

Physiological and Pathological Implications

Role in homeostasis and aging

The cellular stress response is integral to maintaining homeostasis during embryonic development, where heat shock proteins (HSPs) serve as early protective mechanisms against proteotoxic stress. HSPs, particularly the 70-kDa family (Hsp70s), are among the first proteins synthesized in mammalian embryos, facilitating proper folding and preventing aggregation of nascent proteins essential for cell differentiation and organogenesis. For example, Hsp70 expression is dynamically regulated during embryogenesis to support cell survival under fluctuating environmental conditions, such as temperature variations in utero. This role underscores how stress pathways ensure developmental stability by buffering against intrinsic and extrinsic perturbations. In mature organisms, stress responses enable physiological adaptations to demands like exercise, promoting tissue resilience without pathological consequences. During intense physical activity, activates the and pathways to mitigate damage from (ROS) and mechanical strain, leading to upregulated expression of HSPs and antioxidant enzymes. Short-term intense exercise training, for instance, enhances protein stability and reduces markers of cellular damage, fostering an adaptive environment that improves and . These adaptations exemplify how transient stress signaling maintains metabolic by optimizing energy production and repair processes in response to routine physiological challenges. In the context of aging, the hormesis paradigm illustrates how low-level stressors can bolster longevity by priming endogenous defense networks, including sirtuins and FOXO transcription factors, which coordinate antioxidant and repair functions. Mild oxidative or proteotoxic stress induces hormetic effects that activate SIRT1 and FOXO3, enhancing mitochondrial function and DNA repair to counteract age-related decline. This adaptive strengthening of stress responses has been observed across model organisms, where it promotes healthier aging by amplifying vitagenes like heme oxygenase-1. Caloric restriction, a classic hormetic intervention, engages the integrated stress response (ISR) via eIF2α phosphorylation, redirecting translation toward stress-protective proteins and extending lifespan by 20-50% in rodents and nematodes through improved proteostasis. Despite these benefits, chronic stress response activation imposes trade-offs, often culminating in cellular senescence—a stable proliferative arrest that limits further damage but accumulates with age. Persistent ISR or unfolded protein response signaling can drive senescence by sustaining DNA damage checkpoints and inflammatory secretomes, as seen in models of prolonged oxidative insult. Recent studies highlight how unchecked chronic stress exacerbates senescence in fibroblasts and neurons, contributing to tissue dysfunction. Age-related decline in NRF2 activity reduces transcription of antioxidant genes like NQO1 and GCLC, impairing redox homeostasis and amplifying vulnerability to cumulative stress, as observed in human and murine tissues. This NRF2 attenuation represents a key age-related shift that diminishes the efficacy of protective responses.

Involvement in cancer and neurodegeneration

The cellular stress response plays a dual role in cancer, where dysregulated activation can promote tumorigenesis and therapy resistance while also offering opportunities for selective targeting. Overexpression of heat shock proteins (HSPs), such as and , is frequently observed in various cancers, enabling tumor cells to withstand proteotoxic stress and evade induced by chemotherapeutic agents. For instance, elevated HSP levels stabilize oncogenic proteins and inhibit pro-apoptotic pathways, contributing to chemotherapy resistance in and cancers. Similarly, mutations in the NRF2 pathway, a key regulator of response, drive oncogenesis by conferring constitutive activation that enhances antioxidant defenses and metabolic reprogramming, as seen in lung squamous cell carcinomas where somatic NRF2 mutations impair binding, leading to unchecked and survival advantages. In neurodegeneration, failure of the unfolded protein response (UPR) and (HSR) exacerbates proteotoxicity, particularly in protein aggregation disorders. In , chronic ER stress from tau hyperphosphorylation overwhelms the UPR, promoting tau aggregation into neurofibrillary tangles and synaptic dysfunction, with impaired PERK-eIF2α signaling failing to restore . Likewise, in , α-synuclein accumulation disrupts UPR activation in neurons, leading to ER stress propagation and formation, as evidenced by reduced ATF6 processing and XBP1 splicing in affected brain regions. Genetic evidence underscores this vulnerability: HSF1 knockout in models accelerates amyloid-β oligomerization and , highlighting HSF1's neuroprotective role in clearing misfolded proteins via enhanced chaperone activity. This dysregulation creates a therapeutic window in cancer, where controlled stress induction can overwhelm tumor cells' adaptive capacity without harming normal tissues, contrasting with neuroprotective strategies in neurodegeneration. For example, mitogenic overstimulation combined with stress pathway inhibition exploits cancer cells' reliance on proteostasis networks to induce lethal proteotoxicity. Recent advances post-2020 include modulation of the integrated stress response (ISR) pathway, with inhibitors targeting PERK and GCN2 showing promise in preclinical glioblastoma models by sensitizing tumors to radiotherapy and disrupting adaptive survival under hypoxia. Ongoing efforts, such as those evaluating IRE1 inhibitors, aim to translate these into clinical settings for enhanced therapeutic efficacy.

Therapeutic and Research Applications

Modulating stress pathways for therapy

Modulating cellular stress pathways has emerged as a promising therapeutic to exploit dysregulated stress responses in diseases such as cancer and neurodegeneration, where these pathways contribute to cell survival under adverse conditions. Pharmacological agents targeting key components of stress responses can either inhibit protective mechanisms in malignant cells or enhance adaptive responses in vulnerable neurons, thereby shifting the balance toward disease . Heat shock protein 90 (HSP90) inhibitors, such as geldanamycin and its derivatives, have been developed to disrupt and stability in cancer cells, where HSP90 chaperones overexpressed oncoproteins like mutated kinases and receptors. By binding to the ATP-binding site of , these inhibitors induce proteotoxic stress, leading to ubiquitination and degradation of client proteins, which halts tumor and promotes . Clinical applications include geldanamycin analogs like 17-AAG (tanespimycin), which demonstrated antitumor efficacy in phase II trials for and , though limits broader use. Ongoing research focuses on next-generation inhibitors with improved selectivity to minimize off-target effects in non-cancerous tissues. For neurodegenerative disorders, activators of the nuclear factor erythroid 2-related factor 2 (NRF2) pathway, such as derived from , enhance antioxidant defenses and mitigate . modifies , allowing NRF2 nuclear translocation and upregulation of genes encoding synthesis enzymes and phase II detoxifiers, which reduces neuronal damage in models of Parkinson's and Alzheimer's diseases. Preclinical studies show attenuates microglial activation and , preserving neurons in rotenone-induced models. These findings support 's potential as an adjunct therapy, with phase II trials exploring its efficacy in slowing cognitive decline, though challenges persist. Integrated stress response (ISR) modulators like target the of eukaryotic initiation factor 2 alpha (eIF2α) to restore protein synthesis impaired by chronic stress in diseases and related neurodegeneration. inhibits the downstream effects of eIF2α without altering its levels, thereby alleviating translational repression and formation in affected neurons. In -infected mouse models, administration extended survival and reduced synaptic loss by partially restoring protein synthesis rates. This approach highlights ISR modulation's therapeutic window, as low-level ISR inhibition preserves adaptive responses while countering pathological overactivation. Activation of heat shock factor 1 (HSF1) represents another avenue, particularly for (ALS), where impaired heat shock responses exacerbate . Compounds like arimoclomol prolong HSF1 activation under stress, boosting HSP expression to enhance in motor neurons. A phase trial of arimoclomol in early-stage ALS patients demonstrated safety and trends toward slowed functional decline, though a subsequent phase III study did not meet primary endpoints. Emerging agents, such as M102—a dual NRF2/HSF1 activator—are advancing to phase I trials in 2025, building on preclinical in ALS models. Combination therapies further leverage stress pathway modulation by sensitizing tumors to proteotoxic overload, as seen with proteasome inhibitors like paired with antagonists. inhibition accumulates misfolded proteins, amplifying unfolded protein response (UPR) stress, while blockade impairs refolding capacity, synergistically inducing in resistant cancers such as . This strategy exploits cancer cells' reliance on heightened , with clinical data showing enhanced response rates in relapsed patients compared to monotherapy. Such combinations underscore the value of multi-pathway targeting to overcome adaptive resistance in stress-responsive malignancies.

Biomarkers and emerging discoveries

Biomarkers of the cellular stress response provide valuable tools for diagnosing and monitoring stress-related pathologies. Circulating levels of 70 (HSP70) have been identified as a promising noninvasive for cardiovascular risk, with elevated serum concentrations correlating with the severity of , vascular calcification, and acute onset. Similarly, assays measuring NRF2 signaling activity, such as those evaluating nuclear translocation or downstream like NQO1, enable the assessment of in diseases including and neurodegenerative disorders, where NRF2 dysregulation exacerbates and tissue damage. Recent discoveries have expanded understanding of stress response dynamics beyond traditional pathways. In 2025, assemblysomes were characterized as novel intracellular RNA-protein complexes that function as dynamic hubs within , facilitating mRNA sequestration and translational repression during acute cellular insults like oxidative or . These structures integrate RNA-binding proteins and non-coding RNAs to modulate , offering new insights into stress granule maturation and resolution. Advancements in the integrated stress response (ISR) have highlighted its therapeutic potential, particularly through potentiation strategies for antiviral applications. Optogenetics-based screening in 2025 identified small-molecule ISR potentiators that enhance eIF2α phosphorylation, leading to broad-spectrum antiviral effects against viruses such as herpes simplex virus type 1 and Zika in vitro and in vivo models, without inducing cytotoxicity.00690-7) This approach exploits ISR-mediated translational shutdown to inhibit viral replication across diverse pathogens. Emerging research also addresses environmental stressors linked to . A 2024 review synthesized evidence on how prolonged heat exposure and activate cellular stress responses, impairing immune function and barrier integrity through mechanisms like HSP and NRF2 pathway overload, thereby increasing to infections and chronic diseases in vulnerable populations.00119-2/fulltext)

References

  1. [1]
    Cellular Stress Responses: Cell Survival and Cell Death - PMC
    Cells can respond to stress in various ways ranging from the activation of survival pathways to the initiation of cell death that eventually eliminates damaged ...
  2. [2]
  3. [3]
    MOLECULAR AND EVOLUTIONARY BASIS OF THE CELLULAR ...
    Mar 17, 2005 · The cellular stress response is a universal mechanism of extraordinary physiological/pathophysiological significance. It represents a defense reaction of cells ...
  4. [4]
    Cellular Stress Response - an overview | ScienceDirect Topics
    The cellular stress response (CSR) is a physiological reaction of the cell aiming to counteract the insult, cope with stress conditions, and repair damage.
  5. [5]
    A new puffing pattern induced by temperature shock and DNP in ...
    A new puffing pattern induced by temperature shock and DNP in drosophila. Kurze Mitteilungen; Published: 01 December 1962. Volume 18, pages 571–573, (1962) ...
  6. [6]
    Exploring Stressors: Impact on Cellular Organelles and Implications ...
    Cellular stressors can be broadly categorized into four main types: environmental, chemical, biological, and physical stressors [4]. These stressors encompass ...Missing: classification | Show results with:classification
  7. [7]
    How do cells respond to their thermal environment? - PubMed
    A more severe heat shock increases protein denaturation proportionately and leads to aggregation of both denatured and native proteins. This results in ...
  8. [8]
    The roles of metallothioneins in carcinogenesis
    Aug 23, 2018 · MTs have a high affinity for heavy metals, which means that they can bind to xenobiotic heavy metals to provide protection against metal ...
  9. [9]
    Oxidative Stress: Harms and Benefits for Human Health - PMC - NIH
    Oxidative stress is a phenomenon caused by an imbalance between production and accumulation of oxygen reactive species (ROS) in cells and tissues.
  10. [10]
    Effects of acute and chronic oxidative stress on the blood–brain ...
    May 12, 2022 · Chronic and acute oxidative stress modulate cell turnover and immune cell adhesion. We validated RNA sequencing studies by measuring cell ...
  11. [11]
    Contrasting effects of acute and chronic stress on the transcriptome ...
    Chronic stress induced pronounced transcriptional differences, while acute stress caused few lasting transcriptional effects. However, both acute and chronic ...
  12. [12]
    Advances in the structures, mechanisms and targeting of molecular ...
    Mar 12, 2025 · 1). As a co-chaperone of HSP90, HSP70 functions early in protein folding and detects unfolded or misfolded proteins. The co-chaperone HOP (HSP ...
  13. [13]
    Navigating the landscape of protein folding and proteostasis - Nature
    Oct 23, 2025 · Hsp70 displaces small heat shock proteins from aggregates to initiate protein refolding. EMBO J. 36, 783–796 (2017). Article CAS PubMed ...<|separator|>
  14. [14]
    Cochaperones enable Hsp70 to use ATP energy to stabilize native ...
    Sep 4, 2018 · The heat shock protein 70 (Hsp70) chaperones, vital to the proper folding of proteins inside cells, consume ATP and require cochaperones in assisting protein ...
  15. [15]
    Targeted protein degradation: mechanisms, strategies and application
    Apr 4, 2022 · Protein degradation via the ubiquitin-proteasome system (UPS). Proteins undergo ubiquitin-dependent degradation by a suite of three enzymes.
  16. [16]
    Autophagy modulation as a potential therapeutic target for diverse ...
    Aug 31, 2012 · This article provides an overview of the mechanisms and regulation of autophagy, the role of this pathway in disease and strategies for therapeutic modulation.
  17. [17]
    Molecular mechanism of thermosensory function of human heat ...
    Jan 19, 2016 · In the activated state Hsf1 forms trimers or higher order oligomers and binds to its response elements in heat shock gene promoters (Clos et al.Missing: seminal | Show results with:seminal
  18. [18]
    Feedback regulation of heat shock factor 1 (Hsf1) activity by Hsp70 ...
    Despite over 40 years of intense research, the mechanism of Hsf1 activity regulation remains poorly understood at the molecular level.
  19. [19]
    Signal amplification in the KEAP1-NRF2-ARE antioxidant response ...
    The KEAP1-NRF2-ARE signaling pathway plays a central role in mediating the adaptive cellular stress response to oxidative and electrophilic chemicals.
  20. [20]
    Endoplasmic reticulum stress: molecular mechanism and ... - Nature
    Sep 15, 2023 · ER stress is activated when proteostasis is broken with an accumulation of misfolded and unfolded proteins in the ER.
  21. [21]
    The integrated stress response - PMC - PubMed Central - NIH
    Here, we outline current knowledge of the molecular mechanisms of the ISR, highlighting the importance of eIF2α phosphorylation/dephosphorylation and how ...
  22. [22]
    Translational induction of ATF4 during integrated stress response ...
    Sep 16, 2020 · This discovery provides insights into how ATF4 translation occurs efficiently in stressed cells, where the canonical Met-tRNAiMet delivery ...
  23. [23]
    The integrated stress response in neurodegenerative diseases
    Feb 19, 2025 · This review outlines ISR mechanisms, and its relevance in CNS homeostasis and in neurodegenerative disorders where ER stress and ISR link ...
  24. [24]
    Signal Transduction Pathways Leading to Heat Shock Transcription
    HSP genes are regulated at the transcriptional level by heat shock transcription factor 1 (HSF1) that is activated by stress and binds to heat shock elements ...Missing: seminal | Show results with:seminal
  25. [25]
    Repression of Heat Shock Transcription Factor HSF1 Activation by ...
    Heat shock and other proteotoxic stresses cause accumulation of nonnative proteins that trigger activation of heat shock protein (Hsp) genes.Missing: seminal | Show results with:seminal
  26. [26]
    Sensing the Heat Stress by Mammalian Cells - BMC Biophysics
    Aug 11, 2011 · For T = 42°C the stress is transitioning from mild to severe whereas at T = 44°C it is extremely severe. At any temperature, there is thus a ...Missing: threshold | Show results with:threshold
  27. [27]
    Roles of Heat Shock Proteins in Apoptosis, Oxidative Stress, Human ...
    Heat shock proteins (HSPs) play cytoprotective activities under pathological conditions through the initiation of protein folding, repair, refolding of ...
  28. [28]
    Dual role of heat shock proteins as regulators of apoptosis ... - PubMed
    The cytoprotective functions of HSPs can largely be explained by their anti-apoptotic properties. HSP70 and HSP90 can directly interact with different proteins ...
  29. [29]
    Heat Shock Preconditioning Reduces Ischemic Tissue Necrosis by ...
    Recent experimental studies have elucidated that the induction of HSPs is capable of reducing ischemic necrosis in myocardial, neuronal, and renal tissue.Missing: mild | Show results with:mild
  30. [30]
    Disrupted HSF1 regulation in normal and exceptional brain aging
    Sep 14, 2023 · Thus, stress responses may differ between organs and depend upon regenerative capacity and ability to manage damaged proteins and proteotoxicity ...
  31. [31]
    HSF1 Activation Mechanisms, Disease Roles, and Small Molecule ...
    The stress-responsive kinase DYRK2 activates heat shock factor 1 promoting resistance to proteotoxic stress. Cell death and differentiation. 2021;28:1563-78.
  32. [32]
    Reactive oxygen species and mitochondria: A nexus of cellular ...
    The main sources of cellular ROS are mitochondria and NADPH oxidases (NOXs). In contrast to NOX-generated ROS, ROS produced in the mitochondria (mtROS) were ...3. Mitochondrial Ros In The... · 4. Mitochondrial Ros... · 8. Ros And Nlrp3...
  33. [33]
    KEAP1, a cysteine-based sensor and a drug target for the ... - PMC
    Jun 24, 2020 · Chemical modification of specific cysteine sensors of KEAP1 results in loss of NRF2-repressor function and alterations in the expression of NRF2 ...
  34. [34]
    Role of Nrf2 in Oxidative Stress and Toxicity - PMC - PubMed Central
    This review discusses the impact of Nrf2 on oxidative stress and toxicity and how Nrf2 senses oxidants and regulates antioxidant defense.
  35. [35]
    The Central Role of Cytochrome P450 in Xenobiotic Metabolism—A ...
    Cytochrome P450s comprise the major Phase I family of enzymes capable of catalyzing the oxidative biotransformation of a vast majority of lipophilic xenobiotics ...
  36. [36]
    Molecular Mechanism of the UDP-Glucuronosyltransferase 2B20 ...
    Nov 18, 2020 · UGTs, as significant major phase II drug-metabolizing enzymes, play important roles in the detoxification process (Bock, 2003). Many UGT genes ...
  37. [37]
    Induction of Metallothionein I by Arsenic via Metal-activated ...
    Abstract. Metal-activated transcription factor 1 (MTF1) mediates the induction of metallothioneins I and II by zinc and stress signals.
  38. [38]
    Reactive Oxygen Species‐Induced Lipid Peroxidation in Apoptosis ...
    Oct 13, 2019 · Reactive oxygen species- (ROS-) induced lipid peroxidation plays a critical role in cell death including apoptosis, autophagy, and ferroptosis.Generation of ROS and... · Production of Lipid... · Roles of Lipid Peroxidation in...
  39. [39]
    The impact of airborne particulate matter-based pollution on the ...
    Dec 1, 2024 · This review also highlights how the inhalation of PM can lead to deleterious chronic oxidative stress persisting in the lung post-exposure.
  40. [40]
    Systematic and comprehensive insights into HIF-1 stabilization ...
    Jan 6, 2025 · The review provides a comprehensive insight into HIF-1's regulatory mechanism and paves the way for future research and therapeutic development.
  41. [41]
    Hypoxia-Induced Angiogenesis: Good and Evil - PMC
    Hypoxia-inducible factor (HIF)-1 alpha directly enhances the transcriptional activity of stem cell factor (SCF) in response to hypoxia and epidermal growth ...Missing: paper | Show results with:paper
  42. [42]
    Hypoxia-Inducible Factor (HIF)-1 Regulatory Pathway and its ... - NIH
    This review focuses on the potential of the HIF-1 pathway in therapeutic intervention for the treatment of diseases such as cancer and ischemia.
  43. [43]
    Understanding the Unfolded Protein Response (UPR) Pathway
    Mar 1, 2024 · The UPR is regulated by three ER stress sensors: PERK, ATF6, and IRE1. Normally, these sensors are inactivated in the ER due to their ...
  44. [44]
    Mediators of endoplasmic reticulum stress-induced apoptosis - PMC
    So far, UPR-mediated activation of CHOP and JNK has been shown to be involved. CHOP is known to repress BCL2 gene expression, which increases the proportion of ...Missing: unresolved paper
  45. [45]
    The C/EBP Homologous Protein (CHOP) Transcription Factor ...
    The CHOP pathway plays an important role in ER stress-induced apoptosis due to pathogenic microbial infections, neurological diseases and neoplastic diseases.Missing: unresolved paper
  46. [46]
    Role of Assemblysomes in Cellular Stress Responses - PMC - NIH
    Mar 20, 2025 · Assemblysomes are recently discovered intracellular RNA–protein complexes that play important roles in cellular stress response, regulation of gene expression,Missing: UPR ISR
  47. [47]
    The role of heat shock proteins in reproduction - PubMed
    HSP production is enhanced during in-vitro embryo culture and they are among the first proteins produced during mammalian embryo growth.
  48. [48]
    Hsp70 expression and function during embryogenesis - PMC - NIH
    This review focuses on the expression and function of 70-kDa heat shock proteins (Hsp70s) during mammalian embryogenesis, though many features of ...
  49. [49]
    Heat Shock Proteins and Their Role in Pregnancy - PMC - NIH
    Moreover, HSPs are amongst the first proteins synthesized during the development of the mammalian embryo. They affect almost all stages of reproduction (Neuer ...
  50. [50]
    The exercise-induced stress response of skeletal muscle ... - PubMed
    Skeletal muscle adapts to the stress of contractile activity via changes in gene expression to yield an increased content of a family of highly conserved ...
  51. [51]
    Short-term intense exercise training reduces stress markers and ...
    Collectively, these data suggest that short-term intense training enhances protein stability, creating a cellular environment capable of resistance to exercise- ...
  52. [52]
    Exercise-Induced Oxidative Stress: Cellular Mechanisms and Impact ...
    It is now well established that both resting and contracting skeletal muscles produce reactive oxygen species and reactive nitrogen species.
  53. [53]
    Cellular Stress Responses, The Hormesis Paradigm, and Vitagenes
    This article describes in mechanistic detail how hormetic dose responses are mediated for endogenous cellular defense pathways.
  54. [54]
    Cellular stress responses, hormetic phytochemicals and vitagenes ...
    Accordingly, it is proposed that the signaling cascades mediated via Sirtuins and FoxO represent the life-span extending, anti-aging type of regulation. Effects ...
  55. [55]
    Calorie restriction modulates the transcription of genes related to ...
    Calorie restriction (CR), the most‐studied non‐genetic intervention to counter the effects of aging, increases health span and longevity in most model organisms ...2. Results · Table 2 · 4. Methods
  56. [56]
    The mitochondrial integrated stress response: A novel approach to ...
    The ISR is a cellular signaling pathway that responds to various physiological changes and types of stimulation. The mitochondrial integrated stress ...
  57. [57]
    Mechanisms of Cellular Senescence: Cell Cycle Arrest ... - Frontiers
    Constitutive DNA damage response (DDR) signaling leads to chronic activation of p53 which induces cellular senescence. Inactivation of p53 mediated signaling by ...Introduction · Senescence Mediated Cell... · The Senescence Associated...
  58. [58]
    NRF2 in age-related musculoskeletal diseases: Role and treatment ...
    Studies have demonstrated a decline in NRF2 expression in aging cells compared with younger cells, particularly in fibroblasts.
  59. [59]
    The Nrf2-HO-1 system and inflammaging - Frontiers
    Increased oxidative stress is also a key factor associated with aging and there is convincing evidence that Nrf2, not only declines with age, but that Nrf2 and ...
  60. [60]
    Geldanamycin, a Naturally Occurring Inhibitor of Hsp90 and a Lead ...
    Oct 3, 2024 · As discussed, in cancer cells, Hsp90 can serve to prevent the misfolding or degradation of numerous overexpressed or mutated oncoproteins, and ...Introduction · 17-Substituted Derivatives of... · 19-Substituted Derivatives of...
  61. [61]
    Molecular basis for the actions of Hsp90 inhibitors and cancer therapy
    Aug 3, 2011 · Hsp90 inhibitor potently inhibited cellular proliferation by inducing G1 arrest in a variety of tumor cell lines. The G1 arrest by geldanamycin ...Hsp90 Inhibitors And... · Hsp90 Inhibitors Are... · Hsp90 Inhibitors In Various...
  62. [62]
    Potent Hsp90 Inhibitors with Significant Potential in Cancer Therapy
    Oct 20, 2024 · Geldanamycin is a potent heat shock protein inhibitor with remarkable antiproliferative activity. However, it shows pronounced hepatotoxicity in animal models.
  63. [63]
    An update on the status of HSP90 inhibitors in cancer clinical trials
    Notably, improved clinical outcomes have been observed when HSP90i are used in combination therapies, as they exhibit a synergistic antitumor effect. However, ...An Update On The Status Of... · Classification Of Hsp90i... · Hsp90 C-Terminal Domain...
  64. [64]
    The neuroprotective mechanisms and effects of sulforaphane - PMC
    Jun 27, 2019 · SFN is a powerful antioxidant and anti-inflammatory phytochemical with great promise in its ability to protect the nervous system from many diseases and toxins.
  65. [65]
    NRF2 as a Therapeutic Target in Neurodegenerative Diseases
    Sulforaphane activates NRF2 through direct electrophilic modification of the cysteines on KEAP1 allowing for dissociation of NRF2 and nuclear translocation ( ...
  66. [66]
    Sulforaphane protects against rotenone-induced neurotoxicity in vivo
    Aug 24, 2016 · Collectively, treatment with sulforaphane rescued rotenone induced oxidative damage by activating the Nrf2 pathway. Sulforaphane inhibited ...<|separator|>
  67. [67]
    Sulforaphane as a Potential Protective Phytochemical against ...
    Sulforaphane appears to be a promising compound with neuroprotective properties that may play an important role in preventing neurodegeneration.
  68. [68]
    The small molecule ISRIB reverses the effects of eIF2α ... - PMC - NIH
    Feb 26, 2015 · By a yet unknown mechanism, ISRIB blunts the effects of eIF2α phosphorylation in cells and thus represents the first bona fide ISR inhibitor ...
  69. [69]
    Small molecule ISRIB suppresses the integrated stress response ...
    Jan 23, 2019 · We demonstrate here that ISRIB inhibits low-level ISR activity, but does not affect strong ISR signaling.
  70. [70]
    Partial restoration of protein synthesis rates by the small molecule ...
    Mar 5, 2015 · We found that treatment with the small molecule ISRIB, which restores translation downstream of eIF2α, conferred neuroprotection in prion-diseased mice without ...
  71. [71]
    Chemical inhibition of the integrated stress response impairs the ...
    Oct 8, 2024 · By preventing the inhibitory effect of phosphorylated eIF2α on eIF2B, ISRIB blunts the ISR's ability to block protein synthesis and induce the ...<|separator|>
  72. [72]
    arimoclomol | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY
    Arimoclomol is reported to prolong the activation of heat shock transcription factor 1 (HSF1; Q00613), the master regulator of stress-inducible Hsps, including ...
  73. [73]
    Safety and efficacy of arimoclomol in patients with early amyotrophic ...
    A randomised, placebo-controlled, phase 2 trial involving 84 patients with amyotrophic lateral sclerosis treated with up to 300 mg/day of arimoclomol over 12 ...Missing: II | Show results with:II
  74. [74]
    Aclipse Therapeutics Selected to Present M102 ALS Clinical ...
    Nov 10, 2024 · M102, an NRF2 and HSF1 activator, to initiate Phase 1 human trials in Q1 2025 ... drug response, and our clinical development plans in ALS.Missing: II | Show results with:II
  75. [75]
    The Proteasome as a Target for Cancer Therapy - AACR Journals
    Proteasome inhibitors induce apoptosis, have in vivo antitumor efficacy, and sensitize malignant cells and tumors to the proapoptotic effects of conventional ...Abstract · Introduction · Molecular Targets of... · Proteasome Inhibitors...
  76. [76]
    Proteotoxic stress targeted therapy (PSTT) - PMC
    The paper by Neznanov et al. [18] offers an alternative approach towards sensitization of cancer cells to proteasome inhibitors. Instead of inhibition of ...
  77. [77]
    Targeting proteostasis for cancer therapy: current advances ...
    Oct 21, 2025 · Integrated stress response (ISR) is a biological response that cells use to cope with various stressors, including nutrient deprivation, ...
  78. [78]
    Proteasome stress sensitizes malignant pleural mesothelioma cells ...
    Dec 15, 2017 · We found that highly sensitive MPM lines display lower proteasome activity than more bortezomib-resistant clones, suggesting that reduced ...
  79. [79]
    The circulating heat shock proteins as a systems biomarker for ...
    Sep 17, 2025 · Heat Shock Proteins (HSPs) have essential roles in cellular stress responses and have emerging clinical relevance in cardiovascular diseases ...Missing: risk | Show results with:risk
  80. [80]
    Biomarkers of NRF2 signalling: Current status and future challenges
    Dysregulation of NRF2 signalling has been linked to several diseases associated with oxidative stress and inflammation, including neurological disorders ...
  81. [81]
    Role of Assemblysomes in Cellular Stress Responses
    Feb 28, 2025 · Assemblysomes are intracellular RNA-protein complexes that play important roles in cellular stress response, gene expression, and protein ...
  82. [82]
    Potentiating the integrated stress response - Nature
    Sep 3, 2025 · In vitro, the ISR potentiators exhibited broad-spectrum antiviral activity, reducing viral levels of herpes simplex virus type 1 (HSV-1) ...