Damage-associated molecular patterns (DAMPs) are endogenous molecules released from damaged, stressed, or dying host cells that act as danger signals, alerting the innate immune system to tissue injury or cellular stress in the absence of infection.[1] These molecules, also known as alarmins, are recognized by pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) and NOD-like receptors (NLRs), thereby initiating inflammatory responses to promote repair and pathogen clearance.[2] The concept of DAMPs emerged from Polly Matzinger's 1994 "danger model" of immunity, which shifted focus from foreignness to danger signals in immune activation.[1]DAMPs constitute a heterogeneous group derived from various cellular compartments, including intracellular sources like the nucleus (e.g., high-mobility group box 1 protein, or HMGB1) and cytosol (e.g., S100 proteins and heat shock proteins, or HSPs), as well as extracellular matrix components (e.g., biglycan and hyaluronan fragments).[1] Upon release during necrosis, apoptosis, or mechanical stress, these molecules undergo conformational changes or modifications that enable their immunogenic activity, distinguishing them from their physiological roles inside intact cells.[3] Common examples include ATP, uric acid, and mitochondrial DNA, which mimic pathogen-associated molecular patterns (PAMPs) to engage similar PRRs.[2]In health, DAMPs facilitate sterile inflammation essential for wound healing and immune surveillance by recruiting immune cells and inducing cytokine production, such as interleukin-1 (IL-1) and tumor necrosis factor (TNF).[1] However, dysregulated DAMP signaling contributes to pathological conditions, including autoimmune diseases (e.g., rheumatoid arthritis via HMGB1-TLR4 interactions), chronic inflammation in atherosclerosis, and cancer progression through tumor-promoting microenvironments.[1] Ongoing research positions DAMPs as biomarkers for injury severity and potential therapeutic targets, with strategies like neutralizing antibodies against specific DAMPs showing promise in preclinical models; recent studies as of 2025 also highlight their roles in trained immunity and prognostic models for sepsis and cancer.[1][4][5]
Introduction
Definition and Characteristics
Damage-associated molecular patterns (DAMPs) are endogenous molecules that are released or exposed upon cellular stress, damage, or death, serving as signals of tissue injury in the absence of infection. These molecules alert the immune system to initiate protective responses, thereby promoting sterile inflammation without the involvement of pathogens. DAMPs originate from various cellular compartments, including intracellular structures such as the mitochondria and nucleus, as well as extracellular matrix components and plasma membrane elements.[6]DAMPs exhibit a highly heterogeneous nature, encompassing diverse chemical classes such as proteins, nucleic acids, lipids, and metabolites, which reflect their broad origins within the host. In their physiological "day job," these molecules typically perform essential homeostatic functions, such as maintaining cellular structure or facilitating intracellular signaling; however, upon relocation to the extracellular space, they acquire an alarm function that triggers immune activation. This shift in context endows DAMPs with immunogenicity, enabling them to stimulate innate immune cells and amplify inflammatory cascades.[6][1]Key characteristics of DAMPs include their capacity to activate innate immunity through interactions with pattern recognition receptors on immune and non-immune cells, leading to the production of pro-inflammatory mediators. They play a central role in orchestrating sterile inflammation, a process that coordinates tissue repair and pathogen defense in response to endogenous threats. Release mechanisms for DAMPs involve both passive leakage from necrotic or pyroptotic cells and active secretion from stressed but viable cells, often via vesicular pathways or autophagy.[1][6]
Distinction from Pathogen-Associated Molecular Patterns
Pathogen-associated molecular patterns (PAMPs) are conserved molecular motifs derived from microorganisms, such as lipopolysaccharide (LPS) from the outer membrane of Gram-negative bacteria and flagellin from bacterial flagella, which serve as hallmarks of infection and are recognized by the host immune system as exogenous threats.[7] In stark contrast, damage-associated molecular patterns (DAMPs) originate from the host's own cells and tissues, becoming immunogenic only when released, modified, or exposed due to cellular stress, injury, or necrosis, thereby signaling endogenous danger without the presence of pathogens.[7] This fundamental difference underscores PAMPs' role as constant indicators of microbial invasion versus DAMPs' conditional activation tied to host damage.Both PAMPs and DAMPs engage the innate immune system through shared pattern recognition receptors (PRRs), including Toll-like receptors (TLRs) and NOD-like receptors (NLRs), yet they elicit responses in divergent physiological contexts.[8] PAMPs primarily drive antimicrobial defenses, such as phagocytosis and production of pro-inflammatory cytokines to eliminate invading pathogens, whereas DAMPs mediate sterile inflammatory responses focused on tissue repair, recruitment of immune cells, and promotion of healing in the absence of infection.[7] For instance, the same PRR like TLR4 can bind LPS to trigger anti-infective pathways or host-derived molecules to initiate damage-specific signaling, highlighting how receptor context and ligand origin modulate immune outcomes.[8]Evolutionarily, the coexistence of PAMPs and DAMPs supports a unified framework for innate immunity, as articulated in Polly Matzinger's "danger model," which shifts emphasis from mere discrimination of self versus non-self to detection of actual threats to tissue integrity.[9] Under this model, PAMPs represent an ancient adaptation for recognizing conserved microbial patterns to mount rapid defenses against invaders, while DAMPs extend this capability to monitor and respond to internal disruptions, fostering a more flexible system that prioritizes host survival over strict foreignness.[9] This integrated approach likely evolved to balance infection control with homeostasis maintenance across diverse environmental challenges.[9]
Historical Development
Early Concepts of Danger Signals
In the 1970s, researchers identified endogenous mediators released by leukocytes that induced fever and inflammation in the absence of infection, laying early groundwork for understanding non-pathogen-driven immune activation. Specifically, Charles Dinarello and colleagues demonstrated the existence of two distinct human leukocytic pyrogens, later recognized as interleukin-1 alpha (IL-1α) and interleukin-1 beta (IL-1β), which were produced by monocytes and macrophages in response to cellular stress or injury. These findings highlighted that the immune system could respond to internal signals from damaged or activated host cells, rather than solely to external microbial threats.[10]The 1980s and 1990s marked a conceptual shift in immunology, challenging the traditional self/non-self discrimination model that dominated the field. In 1989, Charles Janeway proposed the pattern recognition hypothesis, suggesting that the innate immune system relies on germline-encoded receptors to detect conserved molecular patterns associated with pathogens, thereby priming adaptive immunity. This idea emphasized the role of microbial signals in initiating immune responses but also implicitly questioned the exclusivity of foreignness as the trigger, opening the door to considerations of endogenous cues.00242-8)Building on this evolving perspective, Polly Matzinger introduced the danger model in 1994, positing that the immune system discriminates between dangerous and safe entities rather than self and non-self. In this framework, immunity is triggered by "danger signals" emanating from damaged or necrotic cells, which alert antigen-presenting cells to mount a response regardless of the source's origin.[11] Matzinger argued that such signals arise from tissue injury or stress, providing a more unified explanation for immune activation in various contexts.Initial evidence supporting these danger concepts came from observations of robust immune responses in scenarios devoid of pathogens, such as sterile trauma and organ transplantation. For instance, surgical trauma or burns elicited systemic inflammation and cytokine release similar to infectious states, indicating that host-derived injury signals could drive innate and adaptive immunity.[11] Similarly, allograft rejection in transplants occurred even under sterile conditions, suggesting that cellular damage from ischemia or necrosis provided sufficient alarm signals to provoke rejection. These findings underscored the physiological relevance of endogenous danger cues in immune surveillance.
Establishment of the DAMP Framework
The concept of damage-associated molecular patterns (DAMPs) was formally proposed in 2003 by Walter G. Land, who introduced the term to describe endogenous molecules released during tissue injury that serve as equivalents to pathogen-associated molecular patterns (PAMPs) in activating innate immunity, particularly in the context of allograft rejection and oxidative stress.[12] This proposal built on earlier ideas of danger signals but provided a specific nomenclature for host-derived alarm molecules that signal cellular damage without infection.[13]In the mid-2000s, the framework was further consolidated through connections to the danger model of immunity. Seong and Matzinger (2004) explicitly linked DAMPs to this model by identifying hydrophobicity as an ancient DAMP that triggers innate immune responses via pattern recognition receptors, emphasizing their role in distinguishing self-damage from foreign threats. Concurrently, the identification of alarmins—preformed endogenous mediators released upon cell stress—gained traction in the late 1990s and early 2000s, with key examples like high-mobility group box 1 (HMGB1) and S100 proteins recognized as prototypical DAMPs that amplify inflammation.[1]Post-2010 developments expanded the DAMP framework beyond infectious contexts, highlighting their activation in sterile conditions such as trauma, ischemia-reperfusion injury, and autoimmunity, where they drive hyperinflammation and tissue remodeling.[14] Recent updates in 2024–2025 have incorporated metabolite-derived DAMPs, such as succinate and itaconate from dysregulated metabolism, which modulate immune responses in chronic inflammation and metabolic disorders.[15][4]Key publications have integrated DAMPs into broader innate immunity paradigms, including a 2018 review that synthesized their contributions to inflammatory diseases by detailing how DAMPs from necrotic cells perpetuate cycles of immune activation.[1] These works underscore the evolution of DAMPs from a transplant-focused concept to a central pillar of sterile inflammationresearch.
Molecular Mechanisms
Recognition by Receptors
Damage-associated molecular patterns (DAMPs) are recognized by pattern recognition receptors (PRRs) on immune and non-immune cells, initiating innate immune responses to sterile injury. These receptors detect endogenous molecules released or exposed during cellular damage, distinguishing them from pathogen-associated molecular patterns (PAMPs) through structural similarities that allow DAMPs to engage the same sensing machinery.[2]Toll-like receptors (TLRs), a major family of PRRs, are transmembrane proteins that bind DAMPs at cell surfaces or in endosomes. Surface-localized TLRs, such as TLR2 and TLR4, recognize extracellular DAMPs like high-mobility group box 1 (HMGB1) and S100 proteins; for instance, HMGB1 binds TLR4 via its interaction with the MD-2 co-receptor, mimicking lipopolysaccharide from bacteria.[16] Endosomal TLRs, including TLR3, TLR7, and TLR9, detect internalized nucleic acid DAMPs; mitochondrial DNA acts as a DAMP by engaging TLR9, triggering responses akin to bacterial CpG motifs. These interactions often involve multivalent binding, where multiple DAMP molecules cluster to enhance receptor activation and signaling efficiency.[17]The receptor for advanced glycation end products (RAGE), a multiligand cell surface receptor, primarily recognizes protein DAMPs such as HMGB1 and S100 family members on various cell types. HMGB1 binds RAGE through specific box domains, facilitating recognition independent of TLRs in some contexts, while S100 proteins engage RAGE to promote inflammatory cell recruitment.[18]NOD-like receptors (NLRs), cytosolic sensors, detect intracellular DAMPs like uric acid crystals, which activate NLRP3 by inducing potassium efflux and lysosomal damage. Mitochondrial formyl peptides, resembling bacterial N-formylmethionine motifs, bind formyl peptide receptor 1 (FPR1), a G protein-coupled PRR on the cell surface, to elicit chemotaxis and degranulation.RIG-I-like receptors (RLRs), including RIG-I and MDA5, are cytoplasmic RNA helicases that can recognize endogenous RNA DAMPs released during cellular stress or necrosis, such as mitochondrial transcripts, though their primary role involves viral patterns.[19]These PRRs are predominantly expressed on innate immune cells, including macrophages and dendritic cells, enabling rapid detection of DAMPs to coordinate responses, but they are also present on non-immune cells like endothelial and epithelial cells, broadening the scope of DAMP surveillance.[17][2]
Signaling Pathways and Cellular Responses
Upon recognition by pattern recognition receptors such as Toll-like receptors (TLRs), damage-associated molecular patterns (DAMPs) trigger intracellular signaling cascades that orchestrate innate immune activation. These pathways converge on key adaptor proteins and lead to the transcriptional regulation of inflammatory mediators, ensuring a coordinated response to cellular stress or injury.[2]In the TLR-mediated pathways, the adaptor proteins MYD88 and TRIF are pivotal. The MYD88-dependent cascade, utilized by most TLRs, involves recruitment of interleukin-1 receptor-associated kinases (IRAK1/4) and TRAF6, which culminate in the activation of NF-κB and mitogen-activated protein kinases (MAPKs). This activation promotes the translocation of NF-κB to the nucleus and phosphorylation of MAPKs, driving the expression of pro-inflammatory genes.[2] Complementing this, the TRIF-dependent pathway, primarily associated with TLR3 and TLR4 endosomal signaling, engages TBK1 to activate interferon regulatory factor 3 (IRF3), inducing type I interferon responses.[2] For NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation, the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) oligomerizes with NLRP3 and pro-caspase-1, forming the inflammasome complex that autocleaves and activates caspase-1.[20]The downstream outcomes of these cascades include robust production of cytokines and chemokines, such as IL-1β and TNF-α, which amplify inflammation; generation of reactive oxygen species (ROS) via NADPH oxidase to enhance antimicrobial activity; recruitment of neutrophils and monocytes to the damage site; and induction of pyroptosis through caspase-1-mediated cleavage of gasdermin D, releasing additional inflammatory signals.[21] These responses are highly context-dependent: in acute injury, DAMP signaling elicits strong pro-inflammatory effects to promote tissue repair and pathogen clearance, whereas prolonged or chronic exposure shifts toward tolerogenic outcomes, mitigating autoimmunity through regulatory mechanisms like negative feedback loops.[21]DAMP pathways exhibit significant crosstalk with those triggered by pathogen-associated molecular patterns (PAMPs), sharing adaptors like MYD88 and transcription factors such as NF-κB, which synergistically amplify immune activation during combined sterile injury and infection. This integration ensures heightened vigilance but can exacerbate pathology if dysregulated.[22]
Types and Examples
Protein DAMPs
Protein damage-associated molecular patterns (DAMPs) encompass a diverse group of endogenous proteins released from damaged or stressed cells in humans, signaling danger to the immune system. These molecules, often intracellular or extracellular matrix components, adopt proinflammatory conformations upon extracellular exposure, distinct from their physiological roles. Key examples include nuclear proteins like high-mobility group box 1 (HMGB1) and chaperones such as heat shock proteins (HSPs), which highlight the dual functionality of proteins as both cellular maintainers and alarm signals.[23]Intracellular proteins represent a major category of protein DAMPs, primarily originating from the nucleus or cytoplasm. HMGB1 is a 25-kDa nuclear protein with two DNA-binding HMG-box domains and an acidic C-terminal tail, functioning intracellularly as a DNA chaperone that bends and stabilizes chromatin to facilitate transcription and repair. Upon release, its activity becomes redox-dependent: the reduced and disulfide forms promote inflammation via distinct receptor interactions, while the oxidized form is immunologically inert. HMGB1 is passively released from necrotic cells through membrane rupture or actively secreted by stressed immune cells, such as macrophages, via nonclassical vesicular pathways.[24][25][26]Heat shock proteins HSP60 and HSP70 also serve as intracellular protein DAMPs, acting as molecular chaperones that assist in protein folding, prevent aggregation, and maintain cellular homeostasis under stress. HSP60, a 60-kDa mitochondrial protein, forms oligomeric complexes to refold misfolded polypeptides, while HSP70, a 70-kDa cytosolic and nuclear protein, uses ATP-dependent cycles to stabilize nascent chains and target damaged proteins for degradation. Both are released passively during necrosis or actively from viable but stressed cells, such as those exposed to heat or oxidative damage, transitioning from protective intracellular roles to extracellular proinflammatory signals.[27][28][26]Extracellular matrix proteins contribute to protein DAMPs when proteolytically liberated during tissue injury. Biglycan, a small leucine-rich proteoglycan with two glycosaminoglycan chains attached to a core protein, organizes collagenfibrils and regulates matrix assembly in healthy tissues. Upon damage, soluble biglycan is cleaved from the matrix by matrix metalloproteinases and acts as a DAMP, binding Toll-like receptors to initiate inflammation. Similarly, tenascin-C, a large hexameric glycoprotein with epidermal growth factor-like, fibronectin type III, and fibrinogen-like domains, is transiently expressed in remodeling tissues but upregulated and released during injury, where it modulates cell adhesion and promotes proinflammatory cytokine production. Both are primarily released passively through ECM degradation in necrotic environments.[29][30][31][32]Certain cytokines function as protein DAMPs due to their alarmin properties. Interleukin-1α (IL-1α), a 17-kDa precursor cytokine, is constitutively expressed in epithelial and endothelial cells, where the pro-form itself is bioactive and released upon necrosis to amplify local inflammation. IL-1β, processed from a 31-kDa precursor by the inflammasome into a mature 17-kDa form, is stored intracellularly and secreted actively from activated monocytes or passively from dying cells. S100 proteins, such as the heterodimer S100A8/S100A9 (calprotectin), are 10-14 kDa calcium-binding proteins with EF-hand motifs, abundantly expressed in neutrophils and monocytes; they form disulfide-linked complexes extracellularly to enhance proinflammatory signaling. These cytokines are released via necrosis-induced leakage or active exocytosis from stressed phagocytes.[33][34][35][26]The release of protein DAMPs occurs through two primary mechanisms: passive leakage from necrotic or necroptotic cells, where plasma membrane rupture allows intracellular contents to spill into the extracellular space, and active secretion from viable but stressed cells, often involving leaderless pathways like lysosomal exocytosis or extracellular vesicles. These processes ensure rapid detection by pattern recognition receptors such as Toll-like receptors (TLRs) and receptor for advanced glycation end products (RAGE), though detailed signaling is addressed elsewhere.[26][28]
Non-Protein DAMPs
Non-protein damage-associated molecular patterns (DAMPs) encompass a diverse array of endogenous molecules, including nucleic acids, metabolites, and lipids, that signal cellular damage and trigger innate immune responses in humans. These molecules are released during cellular stress, necrosis, or mitochondrial dysfunction, acting as alarm signals to promote inflammation and repair. Unlike protein DAMPs, non-protein variants often derive from metabolic intermediates or structural components, with recent research (2023–2025) highlighting their roles as metabolite-derived signals in sterile inflammation.[36]Nucleic acids serve as prominent non-protein DAMPs, particularly mitochondrial DNA (mtDNA) and RNA, which are liberated from damaged mitochondria or cytosol during stress. mtDNA, resembling bacterial DNA due to its unmethylated CpG motifs, is released via necrosis, pyroptosis, or vesicular exocytosis and activates the cGAS-STING pathway, leading to type I interferon production and antiviral-like responses. This process is implicated in human conditions such as sepsis, cardiovascular diseases, acute kidney injury, and neurodegeneration. Cytosolic DNA, including mtDNA fragments, is sensed by cyclic GMP-AMP synthase (cGAS), which catalyzes the formation of the second messenger cGAMP to activate STING and downstream NF-κB and IRF3 signaling, exacerbating inflammation in liver injury and neuroinflammatory disorders. Extracellular RNA (exRNA), elevated up to 100-fold under stress, engages Toll-like receptors 7 and 8 (TLR7/8) on immune cells, promoting cytokine secretion like IL-6 and IFN-α in psoriasis and atherosclerosis.[36][37][38]Metabolites represent a rapidly emerging class of non-protein DAMPs, with 2024–2025 studies emphasizing their derivation from disrupted metabolic pathways during cellular injury. Extracellular ATP, released from mitochondria amid stress, binds P2X7 receptors on macrophages and dendritic cells, triggering potassium efflux, NLRP3 inflammasome assembly, and IL-1β/IL-18 maturation, which drives pyroptosis in nonalcoholic fatty liver disease, lung fibrosis, and brain injury. Uric acid crystals, formed from purine metabolism, activate NLRP3 via lysosomal destabilization and ROS production, contributing to gouty arthritis, renal inflammation, and atherosclerosis by amplifying IL-1β responses. Succinate, an intermediate of the tricarboxylic acid cycle, accumulates and is extruded from dysfunctional mitochondria, stabilizing hypoxia-inducible factor-1α (HIF-1α) and enhancing NLRP3 activation through succinate receptor 1 (SUCNR1/GPR91) on immune cells; this promotes pro-inflammatory cytokine release in acute pancreatitis and metabolic disorders. These metabolites underscore the link between bioenergetic failure and immune activation in human pathology.[15][39][40][41]Lipids and glycosaminoglycans also function as non-protein DAMPs, often externalized from intracellular compartments during damage. Cardiolipin, a mitochondrial inner membranephospholipid, translocates to the outer membrane under oxidative stress, exposing it to engage TLR4 and activate NF-κB signaling, while also priming NLRP3 for IL-1β production in heart failure and antiphospholipid syndrome. Cholesterol crystals, phagocytosed by macrophages, cause lysosomal rupture and cathepsin B release, thereby igniting NLRP3 inflammasome responses in atherosclerosis and fatty liver disease. Heparan sulfate fragments, generated by enzymatic cleavage of extracellular matrix during tissue injury, bind receptor for advanced glycation end-products (RAGE), inducing NF-κB-mediated chemokine expression and immune cell recruitment in fibrosis and cancer. Mitochondrial release mechanisms, including permeability transition pore opening or vesicular transport, facilitate the efflux of these non-protein DAMPs—such as mtDNA, ATP, and cardiolipin—amplifying systemic inflammation in myocardial infarction, sepsis, and neurodegeneration.[36][42][41]
Plant DAMPs
In plants, damage-associated molecular patterns (DAMPs) are endogenous molecules released or exposed upon cellular damage, such as wounding or herbivory, that alert the immune system to initiate defense responses distinct from those triggered by pathogen-associated molecular patterns (PAMPs). These plant-specific DAMPs often derive from disrupted cell walls or processed proteins and peptides, enabling rapid local and systemic signaling without homologs to animal receptors like RAGE. Unlike animal DAMPs, which frequently involve intracellular proteins released during necrosis, plant DAMPs emphasize extracellular matrix components and hormone-like peptides adapted to sessile lifestyles and vascular transport.30491-8)[43]DAMPs in plants are primarily released through mechanical disruption of cell walls during injury, such as insect feeding or pathogen invasion, leading to fragmentation of structural components and proteolytic processing of precursors. This release facilitates both apoplastic signaling at the damage site and systemic propagation via the vascular system, where mobile DAMPs travel through phloem to distant tissues, amplifying defenses like jasmonic acid biosynthesis. For instance, herbivory-induced wounds expose hidden cell wall epitopes, while enzymes like polygalacturonases cleave polymers into active fragments, ensuring swift immune activation without relying on mobile immune cells.[44][45]Cell wall-derived DAMPs include oligogalacturonides (OGs), short pectin fragments generated by partial degradation of homogalacturonan during wounding. OGs, typically 9-15 galacturonic acid units long, act as potent elicitors by inducing reactive oxygen species production and defense gene expression, as demonstrated in tomato and Arabidopsis. Cellulose fragments, such as cellodextrins, similarly function as DAMPs, triggering pattern-triggered immunity (PTI) through recognition of exposed β-1,4-glucan chains post-injury. These oligosaccharides highlight the plant cell wall's role as a dynamic sensor of integrity breaches.[46][47][43]Peptide and protein DAMPs encompass signaling molecules processed from larger precursors. Systemin, an 18-amino-acid peptide first identified in tomato, is released from phloem-localized prosystemin upon herbivory and systemically induces protease inhibitors and jasmonate signaling. In Arabidopsis, AtPep1 and related peptides derived from PROPEP genes (e.g., PROPEP1-7) are cleaved by metacaspases during stress, amplifying PTI responses like callose deposition. Rapid alkalinization factor (RALF) peptides, cysteine-rich signals present across land plants, also serve as DAMPs in certain contexts, modulating root immunity and extracellular pH to enhance pathogen resistance. These peptides exemplify how plants repurpose developmental signals for defense.[48][49][50]Additional DAMPs include extracellular adenosine triphosphate (eATP), released from damaged cells into the apoplast, where it acts as a central alarm signal triggering calcium influx and mitogen-activated protein kinase cascades. Amino acids like glutamate can similarly function as weak DAMPs under severe damage. A notable recent discovery is the SCOOP (serine-rich endogenous) peptides, identified in 2021 from hydroxyproline-rich glycoproteins; these 13-amino-acid motifs, processed from PROSCOOP precursors, elicit robust immune responses and represent a broad family conserved in seed plants.[51][52][53]Plant DAMPs are recognized by pattern recognition receptors (PRRs), primarily leucine-rich repeat receptor-like kinases (LRR-RLKs) and wall-associated kinases (WAKs) lacking RAGE equivalents. FLS2, known for PAMP perception, co-receives certain peptide DAMPs like AtPep1 via complex formation with co-receptors such as BAK1. EFR, another LRR-RLK, contributes to DAMP sensing in PTI amplification, particularly in bacterial challenge contexts involving damage. Wall-associated kinases, especially WAK1, directly bind OGs and cell wall fragments, initiating downstream signaling for lignin reinforcement. This receptor repertoire ensures precise discrimination of self-damage from external threats.[44][50][54]
Roles in Physiology and Pathology
In Normal Immune Activation and Repair
In the acute phase following tissue injury, damage-associated molecular patterns (DAMPs) play a critical role in initiating a controlled immune response to maintain homeostasis. Released from damaged cells, DAMPs such as high-mobility group box 1 (HMGB1) and adenosine triphosphate (ATP) serve as endogenous danger signals that recruit immune cells, including neutrophils and monocytes, to the site of injury.[55] This recruitment is mediated through interactions with pattern recognition receptors on immune cells, facilitating rapid localization and response to sterile damage. Additionally, DAMPs promote phagocytosis of cellular debris by enhancing the "find-me" signaling for macrophages, which clears necrotic material and prevents secondary complications, thereby supporting efficient debris removal during early repair.[55]During the tissue repair phase, DAMPs contribute to regenerative processes by stimulating angiogenesis and mobilizing stem cells. For instance, HMGB1 induces endothelial cell migration and vascular endothelial growth factor (VEGF) release, promoting new blood vessel formation essential for nutrient delivery and tissue reconstruction. HMGB1 also orchestrates the mobilization of bone marrow-derived mesenchymal stem cells (MSCs) via chemokine receptors like CXCR4, enabling their recruitment to injured sites where they differentiate into reparative cell types. These actions ensure coordinated healing without excessive inflammation, highlighting DAMPs' role in bridging immune activation to restorative outcomes.[55]As inflammation resolves, certain DAMPs facilitate the transition from pro-inflammatory to anti-inflammatory states, aiding in the restoration of tissue homeostasis. ATP, initially pro-inflammatory, is hydrolyzed to adenosine, which acts as a resolving mediator by activating adenosine receptors on immune cells to suppress cytokine production and promote regulatory phenotypes.[55] This switch prevents prolonged immune activation and supports the re-establishment of normal tissue function.From an evolutionary perspective, the DAMP system confers a survival advantage by enabling rapid, adaptive responses to trauma in the absence of infection. Conserved across species, DAMPs trigger innate immune mechanisms that limit further damage and promote healing, enhancing post-injury resilience and overall organismal fitness. This dual functionality underscores their integral role in physiological repair processes.[55]
In Inflammatory and Autoimmune Diseases
Damage-associated molecular patterns (DAMPs) play a pivotal role in driving chronic sterile inflammation in autoimmune and inflammatory diseases by activating innate immune responses without microbial involvement. In these conditions, endogenous molecules released from damaged or stressed cells persistently stimulate pattern recognition receptors, leading to dysregulated cytokineproduction and immune cell recruitment that perpetuate tissue damage.[56]In rheumatoid arthritis (RA), HMGB1 and S100 proteins are key DAMPs contributing to synovial inflammation. HMGB1, released from necrotic synovial cells, binds to receptors such as TLR4 and RAGE on immune cells, promoting the production of pro-inflammatory cytokines like TNF-α and IL-6, which exacerbate joint destruction.[57] Similarly, S100A8/A9 proteins, secreted by activated neutrophils and macrophages in the synovium, amplify inflammation by activating the NLRP3inflammasome and recruiting additional leukocytes, sustaining chronic synovitis.[56]In systemic lupus erythematosus (SLE), nucleic acid DAMPs such as extracellular DNA and RNA fragments drive autoimmunity through impaired clearance of apoptotic cells. These nuclear DAMPs form immune complexes with autoantibodies, activating plasmacytoid dendritic cells via endosomal TLR7 and TLR9, which triggers type I interferon production and B-cell activation, perpetuating the loss of self-tolerance.[58] This process underlies the chronic inflammation in organs like the kidneys and skin.[59]Beyond autoimmune diseases, DAMPs contribute to trauma-induced inflammation, as seen in gout where uric acid crystals act as a DAMP. Soluble uric acid, released from dying cells, activates the NLRP3inflammasome in macrophages, leading to IL-1β secretion and acute joint inflammation characterized by neutrophil influx.[60] In pulmonary conditions like acute respiratory distress syndrome (ARDS), extracellular ATP serves as a DAMP following alveolar injury. ATP signals through P2X7 receptors on immune cells, promoting inflammasome assembly and cytokine release that worsen lungedema and respiratory failure.[61]Pathologically, the persistent release of DAMPs in these diseases fosters autoantibody production by bridging innate and adaptive immunity. Continuous DAMP exposure enhances antigen presentation by dendritic cells, driving T- and B-cell responses that generate autoantibodies, as observed in SLE and RA.[62] Recent advances highlight DAMPs' role in inducing trained immunity, a form of epigenetic reprogramming in myeloid cells that leads to heightened inflammatory responses upon re-exposure. For instance, HMGB1 and other DAMPs promote histone modifications in monocytes, resulting in sustained cytokine hyperproduction that underlies chronicity in autoimmune disorders.[4]Updates from 2024-2025 research emphasize DAMPs' involvement in neuroinflammation following traumatic brain injury (TBI). Mitochondrial DAMPs like HMGB1 and ATP, released from damaged neurons, activate microglia via TLR4 and P2X7, triggering IL-1β and TNF-α release that amplifies secondary braininjury and long-term neurodegeneration.[63] This sterile neuroinflammatory cascade contributes to persistent cognitive deficits in TBI survivors.[64]
In Cancer and Cardiovascular Disorders
Damage-associated molecular patterns (DAMPs) exhibit dual roles in cancer, promoting tumor progression in some contexts while eliciting anti-tumor immunity in others. High-mobility group box 1 (HMGB1), a prototypical DAMP, facilitates pro-tumor effects by enhancing angiogenesis and metastasis; for instance, extracellular HMGB1 stimulates vascular endothelial growth factor (VEGF) expression and endothelial cell migration, thereby supporting neovascularization essential for tumor growth.[65] In gastric cancer models, HMGB1 overexpression via interleukin-8 (IL-8) pathways drives angiogenic responses, contributing to invasive phenotypes.[66] Similarly, HMGB1 promotes epithelial-mesenchymal transition (EMT) and metastatic dissemination in lung and breast cancers by activating signaling cascades such as PI3K/AKT and NF-κB, which upregulate matrix metalloproteinases (MMPs) for extracellular matrix remodeling.[67] These mechanisms underscore HMGB1's contribution to a tumor-permissive microenvironment, where chronic DAMP release from necrotic or stressed cells sustains inflammation and immune evasion.[68]Conversely, DAMPs play a critical anti-tumor role through immunogenic cell death (ICD), a regulated form of apoptosis that exposes DAMPs to prime adaptive immunity. During ICD, surface exposure of calreticulin (CRT) on dying tumor cells acts as an "eat-me" signal, promoting phagocytic uptake by dendritic cells (DCs), while ATP secretion recruits antigen-presenting cells and fosters inflammasome activation for IL-1β release.[69] These DAMPs collectively enhance cross-presentation of tumor antigens to CD8+ T cells, amplifying cytotoxic T-lymphocyte responses against malignancies.[70] A 2025 systematic review highlights how ICD-induced DAMPs, including CRT and ATP, synergize with checkpoint inhibitors to boost T-cell priming in solid tumors, improving therapeutic outcomes in preclinical models.[71] This immunogenicity contrasts with non-immunogenic cell death, emphasizing DAMPs' context-dependent influence on tumor immunity.[72]In cardiovascular disorders, mitochondrial DAMPs (mtDAMPs) drive pathogenesis in atherosclerosis by activating sterile inflammation within plaques. Formyl peptides, derived from mitochondrial N-formylmethionine sequences, bind formyl peptide receptors (FPRs) on immune cells, triggering neutrophil recruitment and reactive oxygen species production that exacerbate endothelial dysfunction and foam cell formation.[73] Oxidized low-density lipoprotein (OxLDL)-induced mitochondrial damage releases these mtDAMPs, linking dyslipidemia to NLRP3inflammasome activation and IL-1β-mediated plaque instability.[74]Cholesterol crystals within atherosclerotic lesions function as particulate DAMPs, piercing phagolysosomes in macrophages to provoke lysosomal rupture, cathepsin B release, and subsequent NLRP3inflammasome assembly, which amplifies local inflammation and promotes plaque rupture.[75] This crystal-induced response sustains a pro-atherogenic environment, as evidenced by elevated crystal deposition correlating with vulnerable plaque phenotypes.[76]HMGB1 further contributes to cardiovascular injury, particularly in ischemia-reperfusion (I/R) scenarios, where its release from damaged cardiomyocytes worsens myocardial infarction. Post-reperfusion, extracellular HMGB1 binds receptor for advanced glycation end-products (RAGE) on cardiac cells, activating NF-κB pathways that heighten neutrophil infiltration, cytokine storms, and apoptosis, thereby expanding infarct size.[77] In diabetic models, the HMGB1-RAGE axis impairs autophagy and exacerbates I/R-induced cardiomyocyte death, highlighting its detrimental role in vulnerable hearts.[78] Recent advances connect DAMPs to inflammaging in cardiovascular disease; a 2024 review delineates how age-related DAMP accumulation, including mtDNA and HMGB1, fosters chronic low-grade inflammation that accelerates endothelial senescence and atherosclerosis progression.[79] This inflammaging paradigm integrates mtDAMPs with vascular aging, offering insights into therapeutic targeting for age-associated CVD.[80]
Clinical and Therapeutic Applications
DAMPs as Biomarkers
Damage-associated molecular patterns (DAMPs) serve as valuable biomarkers for diagnosing and monitoring various diseases due to their release during cellular stress, injury, or death, reflecting underlying tissue damage and inflammatory responses. Circulating levels of specific DAMPs can indicate disease presence and progression, enabling non-invasive assessment through blood or other biofluids. For instance, elevated high-mobility group box 1 (HMGB1) protein in plasma is associated with sepsis diagnosis and severity, as it acts as a late mediator of inflammation released from damaged cells.[81] Similarly, HMGB1 overexpression correlates with poorer prognosis in multiple cancers, including colorectal and breast carcinomas, where higher levels predict reduced survival rates.[82] Another example is S100B, a calcium-binding protein, whose serum concentrations rise in proportion to trauma severity, particularly in traumatic brain injury, aiding in outcome prediction and monitoring neurological damage.[83]The specificity of DAMPs as biomarkers enhances their clinical utility when linked to particular pathological processes, such as mitochondrial dysfunction. Cell-free mitochondrial DNA (mtDNA) serves as a sensitive indicator of mitochondrial damage, with elevated circulating levels observed in cardiovascular diseases (CVD) like heart failure and ischemic conditions, where it reflects oxidative stress and endothelial injury.[84] In acute respiratory distress syndrome (ARDS), mtDNA levels are significantly increased in patients compared to controls, correlating with disease severity and poor outcomes due to its role in amplifying lung inflammation.[85]Recent advances as of 2025 have incorporated artificial intelligence (AI) to develop multi-omics biomarker panels for more accurate diagnosis and risk stratification in inflammatory diseases. Machine learning algorithms integrate multi-omics data, including DAMPs such as mtDNA, with other clinical data to create predictive models for conditions like sepsis, improving sensitivity and specificity over single markers.[86] These AI-driven approaches analyze complex patterns in multi-omics datasets, enabling personalized prognostic assessments in inflammatory contexts.[87] As of November 2025, ongoing research continues to refine these models.Despite their promise, challenges in utilizing DAMPs as biomarkers include their context-dependent release, influenced by factors like tissue type, injury timing, and co-existing conditions, which can lead to variable interpretations. For example, S100B elevations may occur in non-brain traumas, reducing specificity.[88] Additionally, normalization of DAMP levels against baseline or confounding variables, such as age or comorbidities, is essential to avoid false positives, and optimal timing for measurement remains critical due to dynamic release profiles.[89][90]
Strategies for Targeting DAMPs
Therapeutic strategies for targeting damage-associated molecular patterns (DAMPs) primarily focus on modulating their pro-inflammatory effects to treat conditions such as sepsis, shock, and chronic inflammatory diseases, with approaches divided into inhibition to suppress excessive responses and enhancement to bolster anti-tumor immunity. Inhibition strategies aim to neutralize DAMPs or block their receptors, thereby attenuating downstream signaling pathways like NF-κB and TLR4 that drive cytokine storms. Enhancement, conversely, leverages DAMPs to promote immunogenic cell death and immune activation, particularly in oncology. These methods have shown promise in preclinical models, though clinical translation remains challenged by DAMP redundancy and context-dependent roles.[91]Inhibition of DAMPs often employs monoclonal antibodies to directly bind and sequester key molecules. For instance, anti-HMGB1 monoclonal antibodies, such as the chimeric antibody h2g7 or mouse IgG2b clone 2G7, neutralize extracellular HMGB1, reducing its chemoattractant activity and mitigating inflammation in models of arthritis, liver injury, and sepsis. These antibodies prevent HMGB1 binding to receptors like TLR4 and RAGE, thereby decreasing cytokine production such as TNF-α and IL-6. Decoy receptors, including soluble RAGE (sRAGE), act as ligand traps to inhibit DAMP signaling; sRAGE has demonstrated efficacy in blocking HMGB1-induced vascular inflammation and ischemic stroke damage by competing for ligand binding without activating downstream pathways. Small molecule inhibitors provide an alternative, with glycyrrhizin—a triterpene derived from licorice—directly binding HMGB1 to inhibit its release from necrotic cells and suppress its mitogenic effects in sepsis and renal injury models, offering a cost-effective option with established safety in clinical use for other indications.[91][92][93][94][95][96]Enhancement strategies harness DAMPs to amplify immune responses, particularly in cancer immunotherapy where controlled DAMP release promotes antigen presentation and T-cell activation. Extracellular ATP, a prominent DAMP, acts as a "find-me" signal via P2X7 receptors to trigger immunogenic cell death (ICD), enhancing dendritic cell recruitment and anti-tumor efficacy when boosted by chemotherapy or ATP analogs; for example, ATP-mimicking agents have been used to augment ICD in breast cancer models, leading to improved survival through increased calreticulin exposure and HMGB1 release. This approach contrasts with inhibition by exploiting DAMPs' physiological role in alerting the immune system, though it requires precise dosing to avoid systemic inflammation.[70][97]Recent advances from 2024-2025 emphasize multi-targeting tools like anti-DAMP scavengers for acute conditions such as shock and inflammation. Scavenging peptides, such as opsonic peptide 18 (OP18), bind multiple DAMPs including HMGB1 and eCIRP, attenuating gut ischemia-reperfusion injury and lung inflammation in preclinical models by promoting their clearance via phagocytosis. For chronic diseases, targeting DAMP-induced trained immunity—epigenetic and metabolic reprogramming of myeloid cells—has gained traction; strategies include inhibiting glycolysis or mevalonate pathways to reverse hyperinflammatory states in atherosclerosis and rheumatic disorders, with myeloid-specific high-density lipoprotein nanotherapies showing potential to reprogram hematopoietic stem cells and reduce persistent inflammation. These innovations address DAMP heterogeneity by simultaneously neutralizing several molecules or their downstream effects.[93][98][4]Clinical trials targeting DAMPs are ongoing, particularly for rheumatoid arthritis (RA) and sepsis, but face hurdles like specificity and off-target effects due to overlapping DAMP functions in repair and immunity. In RA, NLRP3 inflammasome inhibitors like MCC950, whose Phase II development was halted due to hepatotoxicity, suppress IL-1β release triggered by DAMPs such as S100 proteins, reducing joint inflammation in preclinical models.[36] Anti-HMGB1 antibodies are in exploratory trials to block synovial DAMP signaling. For sepsis, the anti-eNAMPT monoclonal antibody ALT-100, currently in Phase II (NCT05938036), has demonstrated reduced cytokine levels and improved organ function in preclinical models of sepsis and ARDS; as of November 2025, the trial remains ongoing without published interim results.[91] TREM-1 decoy nangibotide completed Phase IIb (ASTONISH trial) with benefits in septic shock by indirectly modulating DAMP responses.[99] Challenges include ensuring tissue-specific delivery to avoid impairing wound healing, with future efforts focusing on combination therapies and biomarkers for patient stratification.[100]