Next Article in Journal
Exploring the Multifaceted Neuroprotective Mechanisms of Bovine Lactoferrin in a Cell Culture Model of Parkinson’s Disease
Previous Article in Journal
Structure-Guided Design of Cyclic Peptide: A Potent Inhibitor Targeting PD-1/PD-L1 Axis with Antitumor Activity
Previous Article in Special Issue
Biological Activities Underlying the Cardiovascular Benefits of Olive Oil Polyphenols: Focus on Antioxidant, Anti-Inflammatory, and Anti-Atherogenic Effects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modulation of Toll-like Receptors with Natural Compounds: A Therapeutic Avenue Against Inflammaging?

Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(23), 11305; https://doi.org/10.3390/ijms262311305
Submission received: 12 October 2025 / Revised: 14 November 2025 / Accepted: 21 November 2025 / Published: 22 November 2025
(This article belongs to the Special Issue Anti-Inflammatory and Anti-Oxidant Effects of Extracts from Plants)

Abstract

Chronic low-grade inflammation, or “inflammaging,” is a defining feature of aging and a key driver of functional decline. Among innate immune sensors, Toll-like receptors (TLRs) are central mediators linking cellular stress to sterile inflammation, yet their modulation in physiological aging remains largely overlooked. This review bridges that gap by integrating molecular and clinical evidence on age-associated TLR remodeling and summarizing preclinical data on natural compounds that suppress TLR signaling. Across diverse inflammatory models, phytochemicals such as curcumin, quercetin, resveratrol, baicalin, and glycyrrhizin consistently downregulate Toll-like receptor 2- (TLR2-), Toll-like receptor 4- (TLR4-), and Toll-like receptor 9- (TLR9-) dependent myeloid differentiation primary response 88 (MyD88)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/mitogen-activated protein kinase (MAPK) pathways, lowering interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor- α (TNF-α) while enhancing IL-10. These mechanisms mirror the molecular signature of inflammaging, supporting TLRs as actionable targets for restoring immune balance. Collectively, the evidence positions natural TLR modulators as a promising, yet untapped, avenue for promoting healthy aging and extending healthspan.

Graphical Abstract

1. Introduction

Inflammation is a fundamental biological response to tissue injury or infection, essential for defense and repair. However, when unresolved, it transitions from an acute, protective process to a chronic, low-grade inflammatory state that progressively damages tissues and accelerates aging [1]. This persistent, subclinical inflammation, termed “inflammaging”, is characterized by modest but sustained elevations of circulating cytokines such as interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and C-reactive protein (CRP), even in the absence of infection [2]. Unlike acute inflammation, inflammaging is sterile and damage-associated, primarily driven by the accumulation of damage-associated molecular patterns (DAMPs) released from stressed, senescent, or necrotic cells [3].
Among the molecular mediators of this process, Toll-like receptors (TLRs) occupy a central position. These pattern-recognition receptors bridge cellular senescence, endogenous danger signals, and the inflammatory cascade [4]. TLRs detect not only pathogen-associated molecular patterns (PAMPs) but also endogenous DAMPs that accumulate with aging—such as mitochondrial DNA (mtDNA), advanced glycation end-products (AGEs), high-mobility group box 1 (HMGB1), and extracellular microRNAs (miRNAs) [5,6,7]. Activation of receptors like TLR2, TLR4, and TLR9 by these self-derived ligands triggers chronic NF-κB and interferon signaling (Figure 1), leading to sustained cytokine production and tissue damage [7,8,9].
Thus, TLRs represent a mechanistic intersection between molecular aging (senescence) and chronic innate immune activation. Importantly, TLR expression is not static over the life course. As summarized in Table 1, several TLRs show age-associated changes in expression or signaling capacity in human tissues and circulating immune cells, potentially amplifying the pro-inflammatory milieu characteristic of aging.
These observations support the notion that age-related shifts in TLR dynamics may serve as both biomarkers and mediators of inflammaging. Evidence from human and experimental studies suggests that overactivation of TLR pathways contributes to inflammaging, while their genetic or pharmacologic inhibition can attenuate age-related inflammation and extend health span [16]. Despite increasing recognition of TLRs as central mediators of innate immune activation, the translational potential of TLR modulation for healthy aging remains largely unexplored. Current research has focused mainly on disease models of acute or chronic inflammation, leaving a gap in understanding whether targeting TLR signaling could mitigate age-related inflammatory drift [17,18]. Only a few studies have examined natural or dietary compounds with TLR-inhibitory activity in the context of healthy aging [19,20]. This review addresses this gap by synthesizing evidence on TLR involvement in inflammaging and compiling data on natural compounds that inhibit TLR pathways in inflammatory contexts. Our aim is to identify mechanistically plausible candidates for future anti-inflammaging interventions, bridging molecular immunology with the emerging field of healthspan extension.

2. TLRs—Viable Targets for Inflammaging

2.1. Genomic and Transcriptomic Evidence in Aging

Accumulating transcriptomic and functional evidence shows that aging profoundly alters innate immune signaling, particularly TLR–mediated pathways [21]. Across multiple human cohorts and cell models, omics analyses consistently reveal shifts in TLR expression or/and downstream responsiveness [10,22]. Aging is characterized by both heightened baseline inflammatory activity and attenuated inducible responses to TLR stimulation [23,24].
The table below (Table 2) summarizes key observational and omics investigations comparing young and elderly populations, highlighting major TLR-related molecular signatures and functional outcomes across tissues and monocyte subsets.
Aging is associated with broad upregulation of pattern-recognition receptor (PRR) genes, most prominently TLR4, reflecting an enhanced innate immune and inflammatory transcriptional program in fibroblasts. Elevated TLR4 expression is linked to cell-cycle gene regulation via TRIF–interferon signaling, while multiple PRRs, including TLR4 and ACE2, are upregulated at the extremes of age, indicating a transcriptional profile primed for heightened innate immune activation [26].
Even within a modest age range (46–68 years), aging is marked by upregulation of innate immune and TLR-related pathways (TLR4, TLR6, TLR9, MyD88, NF-κB), increased expression of oxidative stress and lipid metabolism genes, and reduced mitochondrial and translational activity. Together, these changes define an early transcriptomic signature of immunosenescence and metabolic decline, reflecting systemic immune and metabolic remodeling that underpins low-grade inflammation and reduced physiological resilience [29].
Within immune subsets, classical CD14+CD16 monocytes show high baseline expression of TLR2, TLR4, TLR5, TLR6, and TLR8, as well as MyD88, IL-1β, and other NF-κB–related genes, indicating a primed state for TLR-mediated activation compared with CD16+ subsets. With age, however, TLR-driven responses weaken: while baseline receptor expression remains stable, monocytes from older adults display reduced transcriptional activation and cytokine production following TLR4 and TLR7/8 stimulation, shifting toward oxidative stress and diminished antiviral signaling. These findings position TLR pathway dysregulation as a central mechanism underlying immunosenescence and inflammaging [27].

2.2. Observational Studies: TLR Expression, Inflammation, and Frailty

Correlative studies in human cohorts reinforce the link between TLR activity, systemic inflammation, and age-related frailty. In general, healthy older adults tend to have higher circulating IL-6, TNF-α, and C-reactive protein (CRP) levels than young adults, and those with the highest levels are at greatest risk of morbidity and mortality [30]. TLR upregulation may be one underpinning of this pro-inflammatory milieu [31].
Table 3 summarizes key human observational studies investigating TLR expression and function in relation to frailty, highlighting the study design, analytical methods, TLR targets, and major inflammatory readouts. Together, these findings delineate a pattern of dysregulated TLR signaling—marked by increased basal inflammation, impaired cytokine production, and altered activation phenotypes—that underpins the immune vulnerability of frail older adults.
Aging profoundly reshapes innate immune function, in part through dysregulation of TLR signaling pathways. One key mechanism involves the accumulation of circulating mtDNA, which acts as a persistent endogenous TLR ligand. This continuous stimulation sustains chronic low-grade inflammation, a hallmark of inflammaging, and contributes to frailty and functional decline in older individuals [36].
At the cellular level, aging alters both the composition and activation profile of circulating immune cells. Older adults exhibit an increase in TLR2 expression and in activation markers such as CD86, CD11c, and HLA-DR on monocytes and dendritic cells, alongside a reduction in plasmacytoid dendritic cells (pDCs). In frail individuals, CD40 expression is further elevated on monocytes, reflecting a shift toward a pro-inflammatory phenotype that amplifies basal immune activation and predisposes to chronic inflammation [34].
Aging and frailty are associated with functional impairments in TLR signaling rather than reduced receptor expression. Surface TLR4 levels remained unchanged on monocytes and dendritic cells, but frail older adults showed markedly decreased IL-12p70 and IL-23 production following combined TLR4 and TLR7/8 stimulation. This reduction occurs despite intact proximal signaling pathways (NF-κB, p38, Erk), indicating a downstream defect in cytokine induction. The findings suggest that TLR function is selectively weakened in frailty, leading to diminished Th1/Th17-supporting responses and contributing to the dysregulated innate immunity and immunosenescence characteristic of aging [32].

2.3. Endogenous TLR Agonists (DAMPs) Amplifying Inflammaging

The pro-inflammatory milieu of aging arises partly from lifelong exposure to DAMPs released by damaged or senescent cells. Molecules such as mtDNA, HMGB1, S100 proteins, HSPs, and ATP mimic pathogen signals: debris from apoptotic or necrotic cells activates TLRs or the NLRP3 inflammasome, promoting IL-1β and IL-18 secretion. Over time, accumulation of these sterile inflammatory stimuli contributes to immunosenescence and inflammaging [37,38,39]. Senescent cells are a major DAMP source. They release cytokines and alarmins such as HMGB1 and S100 proteins, perpetuating innate immune activation via chronic TLR engagement [40].

2.3.1. mtDNA

mtDNA, which increases in circulation with age, resembles bacterial DNA due to its unmethylated CpG motifs [37]. When released into the cytosol or extracellular space during stress, it activates the endosomal DNA sensor TLR9. TLR9 signals through MyD88 to induce NF-κB–driven inflammatory genes and IRF7-mediated type I interferons [41].
In older adults, elevated mtDNA levels correlate with systemic inflammation. Individuals with sarcopenia showed higher circulating mtDNA and IL-6/IL-8 levels than controls, with mtDNA independently predicting sarcopenia risk [42]. Other studies link mtDNA to elevated TNF-α and IL-6, reinforcing its role as a pro-inflammatory DAMP [43]. Mechanistically, mtDNA–TLR9 interaction in macrophages and dendritic cells triggers NF-κB activation, inducing IL-1β, IL-6, and TNF-α, and can prime the inflammasome [44]. Chronic TLR9 stimulation in aging tissues also upregulates S100A8/A9, creating a feed-forward loop that sustains inflammation and tissue injury [45].

2.3.2. HMGB1 and TLR4

HMGB1, a nuclear chromatin-binding protein, becomes an extracellular alarmin when released by stressed or dying cells. Its levels rise with age and in disorders such as atherosclerosis and neurodegeneration. HMGB1 activates TLR4 and RAGE on immune cells, engaging MyD88/NF-κB and MAPK pathways that drive TNF-α, IL-1β, and IL-6 production [46]. Clinically, elevated HMGB1 correlates with inflammation and frailty. In elderly surgical patients, higher serum HMGB1 was linked to postoperative delirium and increased IL-6 and IL-1β [47]. Chronic HMGB1 elevation also associates with coronary atherosclerosis and frailty, and HMGB1-rich plasma from aged individuals can induce cellular senescence in vitro [48]. Thus, HMGB1 is a central DAMP in inflammaging, sustaining NF-κB signaling and cytokine release through TLR4/MD-2 activation.

2.3.3. S100 Proteins and TLR Signaling

S100A8 and S100A9 form the heterodimer calprotectin, released by neutrophils, monocytes, and senescent cells under stress. Their levels rise with age and in diseases such as atherosclerosis and Alzheimer’s. S100A8/A9 activate TLR4 and RAGE on immune cells, triggering MyD88–NF-κB and MAPK signaling, which induces IL-6 and TNF-α and promotes a pro-inflammatory macrophage phenotype [49].
Elevated S100A8/A9 correlates with inflammation and functional decline in aging tissues. In elderly individuals with periodontal disease—a model of inflammaging—S100A8/A9 expression is TLR-dependent, and TLR blockade reduces both DAMP and cytokine levels. Thus, S100 proteins likely act as endogenous TLR4 ligands amplifying NF-κB–mediated inflammation in aging [50].

2.3.4. HSPs and Other DAMPs

HSPs are intracellular chaperones that, when released extracellularly under stress, act as DAMPs. In older individuals, extracellular HSP70 and HSP90 activate TLR2 and TLR4 on antigen-presenting cells, inducing NF-κB–dependent cytokines. Elevated HSP70 correlates with IL-6 levels and frailty, linking it to age-related inflammation [38].
Extracellular ATP from dying cells activates the P2X7 receptor, triggering the NLRP3 inflammasome and maturation of IL-1β and IL-18. Though ATP does not bind TLRs directly, it amplifies TLR priming and inflammation. Similarly, uric acid—accumulating with age or in gout—activates TLR2/4 and NLRP3, promoting NF-κB activation and IL-1β release. In older adults with chronic kidney disease, uric acid drives vascular inflammation through TLR4–MyD88–NF-κB and NLRP3 pathways [51].
Other endogenous TLR ligands include extracellular DNA/RNA, histones, and oxidized lipoproteins [52]. Oxidized LDL, common in atherosclerosis, activates macrophage TLR4, triggering NF-κB and inflammasome signaling, while self-RNA can engage TLR7/8, contributing to chronic inflammation and autoimmunity in the elderly [53]. Collectively, age-related accumulation of cellular debris (“garb-aging”) chronically engages TLR2/4 (proteins and lipids), TLR9 (mtDNA), and TLR7/8 (RNA), perpetuating innate immune activation.
Thus, DAMP-activated TLRs on macrophages, dendritic cells, and neutrophils trigger signaling cascades converging on NF-κB and AP-1. Most TLRs use MyD88, activating IRAKs and TRAF6. With age, this pathway becomes chronically active: aged immune cells show exaggerated responses despite feedback inhibition [54].
NF-κB drives key features of inflammaging, inducing SASP cytokines (IL-6, IL-1β, TNF-α, CXCL8) and inflammatory enzymes (iNOS, COX-2). Chronic NF-κB activation underlies many age-related pathologies [55,56]. TLR4 can also signal through TRIF to activate IRF3 and type I interferons, which, though antiviral, can become dysregulated in aging and autoimmunity [57].
TLR signaling also primes the inflammasome by upregulating NLRP3 and pro–IL-1β/IL-18, enabling rapid cytokine release upon secondary signals like ATP. Persistent activation induces parainflammation—a low-grade inflammatory state that damages tissues. Over time, continuous stimulation skews monocytes toward a pro-inflammatory state but can also cause TLR tolerance, reducing responsiveness to new challenges [58]. Thus, chronic DAMP–TLR–MyD88–NF-κB signaling sustains IL-6, IL-1β, and TNF-α production, central to the aging immune phenotype.
The persistent cytokine production is linked to age-associated disease. IL-6, a hallmark of inflammaging, predicts morbidity and mortality [59]. TNF-α, another NF-κB target, promotes muscle wasting [60], insulin resistance [61], and neurodegeneration [62]. IL-1β, generated via inflammasome activation, amplifies IL-6 and acute-phase responses; higher IL-1β expression correlates with frailty in older adults [63]. Thus, these inflammatory cascades contribute to sarcopenia, frailty, and multimorbidity.
At the organ level, DAMP-induced TLR signaling drives pathology across systems: TLR4 activation accelerates atherosclerosis, HMGB1 promotes plaque instability, and microglial TLR4 activation by HMGB1 or S100 proteins fuels neuroinflammation and cognitive decline [64,65]. Older adults with pre-existing inflammaging are also more prone to cytokine storms during infection, as seen in COVID-19 [66].
In summary, collective genomic, transcriptomic, and clinical evidence identifies TLRs as viable therapeutic targets in aging. Aging is marked by chronic upregulation of TLR pathways—particularly TLR2, TLR4, and TLR9—driven by persistent stimulation from endogenous DAMPs such as mtDNA, HMGB1, and S100 proteins. This sustained activation of the MyD88–NF-κB axis promotes low-grade inflammation, cytokine overproduction, and tissue dysfunction, hallmarks of inflammaging and frailty. Conversely, impaired inducible TLR responses in older immune cells contribute to immunosenescence and reduced resilience. Targeting TLR signaling therefore addresses both core mechanisms of aging: chronic inflammation and declining immune adaptability.

3. Natural Modulators of TLRs

Various plant-derived natural compounds attenuate the inflammatory responses by targeting TLR4-mediated pathways [67,68,69]. Experimental evidence from murine and rat models of metabolic, hepatic, renal, and systemic inflammatory disorders demonstrates that phytochemicals such as curcumin, baicalin, paeoniflorin, quercetin, berberine, and others exert anti-inflammatory effects through inhibition of the TLR4/MyD88/NF-κB cascade and its downstream mediators (e.g., MAPK, AP-1, NLRP3) [70,71,72,73,74,75,76,77].
Table 4 below summarizes key studies reporting ex vivo or in vivo effects of these compounds on TLR4-related signaling and cytokine profiles, detailing the experimental models, administration routes, mechanistic targets, and principal outcomes. Collectively, these findings support the concept that natural bioactive substances can modulate TLR4 signaling, mitigating the excessive inflammatory burden implicated in metabolic dysfunction, liver injury, sepsis, and other inflammation-driven diseases.
Across diverse experimental models, these studies consistently demonstrate that natural compounds act as modulators of TLR4-driven inflammation, converging on a few key molecular events:
-
inhibition of canonical TLR4/MyD88/NF-κB signaling. Nearly all compounds (e.g., baicalin, curcumin, berberine, paeoniflorin, quercetin) suppress the MyD88–IRAK–TRAF6 axis, leading to reduced phosphorylation and nuclear translocation of NF-κB p65, and thereby lowering transcription of pro-inflammatory cytokines [70,73,74,77,80].
-
downregulation of MAPK and AP-1 pathways. Several agents concurrently inhibit p38, ERK, and JNK activation, attenuating AP-1 (c-Fos/c-Jun)–dependent transcription [68,71,75,76,88].
-
suppression of downstream inflammatory mediators. The resulting decreases in TNF-α, IL-1β, IL-6, MCP-1, iNOS, and COX-2 are consistent across models of sepsis, liver injury, kidney damage, and metabolic inflammation [76,81,82,84].
-
stabilization of IκB-α and prevention of NF-κB nuclear translocation. This common mechanistic endpoint prevents the persistent transcription of inflammatory genes.
-
interference with TLR4–HMGB1 interaction, observed with paeoniflorin, curcumin, and quercetin, highlighting the blockade of DAMP–TLR4 communication as a recurring anti-inflammatory mechanism [75,79,80].
-
attenuation of oxidative and fibrotic signaling—many compounds reduce oxidative stress markers (NO, iNOS) and fibrogenic mediators (α-SMA, collagen I), linking TLR4 inhibition to improved tissue remodeling [76,79,81,93].
Several naturally derived compounds suppress inflammation by targeting TLR2-dependent signaling pathways, which are key mediators of sterile inflammation and tissue injury. Evidence from murine and rat models of hepatic, renal, pulmonary, and systemic inflammation indicates that phytochemicals such as curcumin, quercetin, paeoniflorin, resveratrol, berberine, and others inhibit the TLR2/MyD88/NF-κB and MAPK cascades, reducing downstream production of pro-inflammatory cytokines (e.g., TNF-α, IL-1β, IL-6) and oxidative stress markers (Table 5) [74,76,95,97,98,99].
Several natural bioactive compounds have emerged as potent regulators of TLR9-dependent inflammatory signaling, particularly in models involving mitochondrial or nuclear DNA–driven sterile inflammation. Studies in rodent models of sepsis, autoimmune, metabolic, and ischemic injury demonstrate that agents such as curcumin, glycyrrhizin, oxymatrine, tanshinone IIA, oleanolic acid, and gastrodin suppress TLR9/MyD88/NF-κB or related pathways (e.g., IRF5/7, JAK2/STAT3) [101,102,103,104,105,106].
These interventions consistently reduce pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-18), oxidative stress markers (MDA, MPO), and tissue damage indicators (NGAL, KIM-1), while enhancing antioxidant and barrier-protective molecules (e.g., SOD, GSH, zonulin-1, occludin) [101,102,103,104,105,106].
Table 6 summarizes key experimental findings highlighting how TLR9 inhibition by natural compounds mitigates DNA-sensing–driven inflammation, oxidative injury, and immune dysregulation—mechanisms that closely mirror the TLR9–mtDNA axis underlying inflammaging in humans.
To better understand the therapeutic potential of natural Toll-like receptor (TLR) modulators, it is essential to consider their chemical diversity, physicochemical characteristics, and safety profiles. These parameters determine not only the molecular interactions with TLRs but also their pharmacokinetic behavior and potential toxicity in vivo. Table 7 summarizes representative natural compounds with reported TLR-modulating activity, organized by chemical class and key molecular descriptors, along with available safety data from experimental and clinical studies. Presenting this information in an integrated format provides a comparative framework for evaluating structure–activity relationships and for identifying promising candidates for further preclinical or clinical development.

4. Discussion

This review addresses a critical gap in current aging research by examining TLRs as mechanistic drivers of inflammaging and as potential therapeutic targets to counteract age-related immune dysregulation. It identifies relevant natural candidates that could be repurposed for anti-inflammaging interventions. The discussion integrates molecular, functional, and translational findings to define how TLR-targeted modulation may restore immune homeostasis and promote resilience in later life.
Older adults exhibit elevated expression of activation markers (CD86, CD11c, HLA-DR) on circulating antigen-presenting cells, higher TLR2 expression on specific monocyte and dendritic cell subsets, and reduced plasmacytoid dendritic cell numbers. Frailty amplifies this primed phenotype through upregulated CD40 expression on monocytes and elevated basal IL-6 levels—defining the pro-inflammatory set point characteristic of inflammaging [29].
Aging profoundly alters innate immune function through dual and seemingly paradoxical trends: baseline activation increases, while inducible responsiveness declines. Older adults display higher expression of activation markers (CD86, CD11c, HLA-DR) on circulating antigen-presenting cells, elevated TLR2 on specific monocyte and dendritic cell subsets, and fewer pDCs. Frailty further amplifies this primed phenotype via upregulated CD40 on monocytes and heightened basal IL-6 levels—defining a pro-inflammatory set-point characteristic of inflammaging [34].
Despite this chronic activation, inducible cytokine responses to TLR ligation are often blunted. In frail elders, TLR4 and TLR7/8 co-stimulation results in reduced IL-12p70 and IL-23 production, despite preserved NF-κB, p38, and Erk activation and unchanged TLR4 surface expression [32]. Multivariate analyses confirm that frailty, dependence, and poor nutrition— rather than chronological age—predict this impairment. Similar patterns of reduced IFN-α, IFN-γ, and IL-1β responses after TLR4/TLR7/8 stimulation indicate weakened Th1/Th17 and antiviral support [32]. In summary, aging manifests as a “primed baseline, smaller fold-change” pattern marked by chronic inflammation but limited adaptability to new immune challenge.
These functional shifts mirror molecular signatures observed in omics studies, which show upregulation of TLR4, TLR6, and TLR9 and their adaptors MYD88 and NFKBIA in older individuals [25,26]. Concomitant elevation of oxidative stress-related genes and diminished mitochondrial activity sustain a feed-forward inflammatory loop. Endogenous DAMPs chronically engage TLRs, activating MyD88/NF-κB signaling and perpetuating IL-6, IL-1β, and TNF-α release.
Thus, TLRs, particularly TLR2, TLR4, and TLR9, represent central molecular hubs linking cellular damage to inflammaging and frailty with TLR4 standing out as a mechanistic bridge between natural anti-inflammatory agents and the molecular hallmarks of inflammaging.
Natural TLR4 modulators may attenuate DAMP-induced TLR4 sustained activation by:
(i)
interrupting DAMP–TLR4 feedback loops,
(ii)
reducing baseline NF-κB activation and IL-6/TNF output that drive frailty and metabolic decline,
(iii)
preserving redox balance to prevent further DAMP release, and
(iv)
limiting macrophage and Kupffer cell hyperactivation, which underlies chronic parainflammation in aging tissues [32,33,34,35,36]. Collectively, these mechanisms support TLR4 inhibition as a translational link between nutritional bioactives and the mitigation of age-related inflammatory pathology.
Evidence from animal and cell models shows that numerous plant-derived bioactive compounds attenuate TLR-mediated inflammatory signaling. Compounds such as curcumin [70], quercetin [80], berberine [77], paeoniflorin [74], glycyrrhizin [99], resveratrol [98], and baicalin [73] suppress MyD88–IRAK–TRAF6–NF-κB and MAPK pathways, thereby reducing IL-6, TNF-α, and IL-1β production. These agents also limit NLRP3 priming and oxidative stress, restore antioxidant defenses, and preserve mitochondrial homeostasis across models of liver injury, neuroinflammation, metabolic dysfunction, and sepsis [94,98]. Several compounds exhibit multi-TLR modulation, suggesting broader anti-inflammatory capacity in complex age-related contexts. Curcumin, glycyrrhizin and quercetin act on TLR2, TLR4, and TLR9 [70,72,76,80,81,102,103]; berberine and paeoniflorin inhibit both TLR2 and TLR4 [74,95]; and genipin interferes with TLR2/TLR9 signaling [96] (Figure 2). Since multiple DAMPs simultaneously activate distinct TLRs during aging, these pleiotropic modulators may outperform single-target inhibitors by disrupting overlapping inflammatory circuits.
Converging transcriptomic, functional, and preclinical data provides a strong rationale for targeting TLR pathways as a geroprotective strategy. Geriatric syndromes such as frailty, sarcopenia, delirium, and dementia share inflammatory underpinnings, with TLR-mediated innate immune activation emerging as a pivotal mechanistic contributor. Elevated TLR signaling through TLR2, TLR4, and TLR9 correlates with frailty, cognitive impairment, and functional decline. Modulating these pathways could recalibrate the inflammatory set point, reducing basal activation without abolishing inducible defenses—a critical distinction in older adults with immune exhaustion.
Natural multi-TLR modulators, with favorable safety profiles and a history of dietary exposure, represent promising candidates for long-term prophylaxis aimed at extending healthspan rather than merely treating disease.
When evaluating TLR modulators as a means to extend healthspan, several limitations should be noted. First, the current evidence is heavily biased toward preclinical or disease-specific contexts, with aging-specific validation remaining sparse [70,77,102]. Thus, direct data linking these compounds to improved healthspan or reduced frailty in humans are limited. Moreover, substantial variation in dose, formulation, and pharmacokinetics further complicates translation with limited information on long-term safety or bioavailability in older populations [45,63]. Most human studies rely on small, heterogeneous cohorts without consistent control for comorbidities, medications, nutritional status, or sex—factors that shape innate immune tone [109]. Chronological age is often conflated with biological aging or frailty, each of which independently affects TLR function. Furthermore, transcriptomic analyses may also reflect changes in immune cell composition rather than intrinsic pathway activation [35,36].
These discrepancies underscore the need for multi-omic, cell-specific, and protein-level analyses to resolve apparent contradictions between TLR expression and functional responsiveness.
The safety of TLR inhibition in older adults also warrants caution. Because immunosenescence already impairs both innate and adaptive responses, further dampening TLR activity could exacerbate susceptibility to infection or reduce vaccine responsiveness [110]. Clinical experience with anti-inflammatory biologics and TLR4 inhibitors demonstrates increased infection risk, unpredictable immune suppression [111], or unpredictable inflammatory rebounds. Broad immune blockade may fail, or even worsen outcomes, outside defined contexts [112].
Moreover, TLRs are crucial for antitumor immunity and tissue repair; chronic inhibition may impair cancer surveillance or wound healing. Therefore, selective or partial modulation—rather than broad inhibition—should be prioritized. Thus, biomarker-guided interventions in DAMP-dominated states—rather than continuous prophylactic use—may allow inflammatory tone to be lowered without compromising essential defenses. Co-optimization with vaccination is likewise crucial to prevent loss of adjuvant benefit. Clinical studies should stratify by frailty, comorbidities, and polypharmacy and include functional immune readouts (ex vivo TLR responsiveness) alongside clinical endpoints.
Finally, it is important to consider the potential for positivity bias in the literature.
Outcomes often varied based on biological context. TLR modulation is highly context-dependent, governed by dose, formulation, target cell type, ligand, and exposure duration. Reports of natural compounds (e.g., curcumin, quercetin, resveratrol) show that inhibition often occurs only at supra-physiological concentrations or with specialized formulations. Conversely, negligible effects are seen at physiological doses. Some studies reported no effect on TLR expression or cytokine release at very low doses, indicating a narrow therapeutic window for reproducible TLR modulation (Table 8). Variability in purity, vehicle (e.g., DMSO, ethanol), and compound chemistry (e.g., glycosides vs. aglycones) further explains inconsistent outcomes. Some agents display biphasic behavior—low-dose priming followed by inhibition at higher doses—highlighting the need for pharmacokinetic realism and standardized protocols in future studies.
Future research should prioritize:
  • Aging-specific translational trials evaluating natural TLR modulators with endpoints such as cytokine profiles (IL-6, IL-1β, TNF-α), circulating DAMPs (mtDNA, HMGB1), and frailty or resilience indices.
  • Mechanistic biomarker development, including ex vivo TLR responsiveness, to assess both basal and inducible immune function.
  • Combination strategies integrating TLR modulation with DAMP-lowering or senolytic interventions.
  • Selective targeting approaches balancing inflammation control with immune competence.
Standardized pharmacokinetic studies, context-specific dosing, and stratification by frailty and comorbidity will be essential to evaluate safety and efficacy. Until such data emerge, natural compounds should be viewed as context-dependent TLR modulators rather than universally effective inhibitors [74,97,98,99].

5. Conclusions

In summary, TLR signaling sits at the intersection of innate immune aging, chronic inflammation, and tissue decline. Natural compounds capable of modulating multiple TLR pathways hold promise as prophylactic agents to rebalance the aged systems. By dampening chronic DAMP–TLR–NF-κB activation while preserving adaptive responsiveness, these agents could offer a feasible route to mitigate inflammaging and promote healthy longevity.

Author Contributions

Conceptualization, A.Z.; methodology, C.P. and A.Z.; data curation, C.P. and G.M.N.; writing—original draft preparation, G.M.N., C.P., and C.A.; writing—review and editing, A.Z.; visualization, C.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A New Immune–Metabolic Viewpoint for Age-Related Diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef] [PubMed]
  2. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic Inflammation in the Etiology of Disease across the Life Span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
  3. Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating Mitochondrial DAMPs Cause Inflammatory Responses to Injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef]
  4. Molteni, M.; Gemma, S.; Rossetti, C. The Role of Toll-Like Receptor 4 in Infectious and Noninfectious Inflammation. Mediat. Inflamm. 2016, 2016, 6978936. [Google Scholar] [CrossRef]
  5. Pascual, M.; Calvo-Rodriguez, M.; Núñez, L.; Villalobos, C.; Ureña, J.; Guerri, C. Toll-like Receptors in Neuroinflammation, Neurodegeneration, and Alcohol-induced Brain Damage. IUBMB Life 2021, 73, 900–915. [Google Scholar] [CrossRef] [PubMed]
  6. Huang, J.; Xie, Y.; Sun, X.; Zeh, H.J.; Kang, R.; Lotze, M.T.; Tang, D. DAMPs, Ageing, and Cancer: The ‘DAMP Hypothesis. ’ Ageing Res. Rev. 2015, 24, 3–16. [Google Scholar] [CrossRef]
  7. Zanini, G.; Selleri, V.; Lopez Domenech, S.; Malerba, M.; Nasi, M.; Mattioli, A.V.; Pinti, M. Mitochondrial DNA as Inflammatory DAMP: A Warning of an Aging Immune System? Biochem. Soc. Trans. 2023, 51, 735–745. [Google Scholar] [CrossRef]
  8. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef]
  9. Duan, T.; Du, Y.; Xing, C.; Wang, H.Y.; Wang, R.-F. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front. Immunol. 2022, 13, 812774. [Google Scholar] [CrossRef]
  10. Panda, A.; Qian, F.; Mohanty, S.; van Duin, D.; Newman, F.K.; Zhang, L.; Chen, S.; Towle, V.; Belshe, R.B.; Fikrig, E.; et al. Age-Associated Decrease in TLR Function in Primary Human Dendritic Cells Predicts Influenza Vaccine Response. J. Immunol. 2010, 184, 2518–2527. [Google Scholar] [CrossRef] [PubMed]
  11. Sridharan, A.; Esposo, M.; Kaushal, K.; Tay, J.; Osann, K.; Agrawal, S.; Gupta, S.; Agrawal, A. Age-Associated Impaired Plasmacytoid Dendritic Cell Functions Lead to Decreased CD4 and CD8 T Cell Immunity. Age 2011, 33, 363–376. [Google Scholar] [CrossRef]
  12. Qian, F.; Guo, X.; Wang, X.; Yuan, X.; Chen, S.; Malawista, S.E.; Bockenstedt, L.K.; Allore, H.G.; Montgomery, R.R. Reduced Bioenergetics and Toll-like Receptor 1 Function in Human Polymorphonuclear Leukocytes in Aging. Aging 2014, 6, 131–139. [Google Scholar] [CrossRef] [PubMed]
  13. Qian, F.; Wang, X.; Zhang, L.; Chen, S.; Piecychna, M.; Allore, H.; Bockenstedt, L.; Malawista, S.; Bucala, R.; Shaw, A.C.; et al. Age-associated Elevation in TLR5 Leads to Increased Inflammatory Responses in the Elderly. Aging Cell 2012, 11, 104–110. [Google Scholar] [CrossRef]
  14. Perkins, R.K.; Lavin, K.M.; Raue, U.; Jemiolo, B.; Trappe, S.W.; Trappe, T.A. Effects of Aging and Lifelong Aerobic Exercise on Expression of Innate Immune Components in Skeletal Muscle of Women. J. Appl. Physiol. 2024, 136, 482–491. [Google Scholar] [CrossRef] [PubMed]
  15. Ghosh, S.; Lertwattanarak, R.; Garduno, J.d.J.; Galeana, J.J.; Li, J.; Zamarripa, F.; Lancaster, J.L.; Mohan, S.; Hussey, S.; Musi, N. Elevated Muscle TLR4 Expression and Metabolic Endotoxemia in Human Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70, 232–246. [Google Scholar] [CrossRef]
  16. Thevaranjan, N.; Puchta, A.; Schulz, C.; Naidoo, A.; Szamosi, J.C.; Verschoor, C.P.; Loukov, D.; Schenck, L.P.; Jury, J.; Foley, K.P.; et al. Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe 2017, 21, 455–466.e4. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, Y.; Zhang, S.; Li, H.; Wang, H.; Zhang, T.; Hutchinson, M.R.; Yin, H.; Wang, X. Small-Molecule Modulators of Toll-like Receptors. Acc. Chem. Res. 2020, 53, 1046–1055. [Google Scholar] [CrossRef]
  18. Gao, W.; Xiong, Y.; Li, Q.; Yang, H. Inhibition of Toll-Like Receptor Signaling as a Promising Therapy for Inflammatory Diseases: A Journey from Molecular to Nano Therapeutics. Front. Physiol. 2017, 8, 508. [Google Scholar] [CrossRef]
  19. Azam, S.; Jakaria, M.; Kim, I.-S.; Kim, J.; Haque, M.E.; Choi, D.-K. Regulation of Toll-Like Receptor (TLR) Signaling Pathway by Polyphenols in the Treatment of Age-Linked Neurodegenerative Diseases: Focus on TLR4 Signaling. Front. Immunol. 2019, 10, 1000. [Google Scholar] [CrossRef]
  20. Chen, C.-Y.; Kao, C.-L.; Liu, C.-M. The Cancer Prevention, Anti-Inflammatory and Anti-Oxidation of Bioactive Phytochemicals Targeting the TLR4 Signaling Pathway. Int. J. Mol. Sci. 2018, 19, 2729. [Google Scholar] [CrossRef]
  21. Shaw, A.C.; Panda, A.; Joshi, S.R.; Qian, F.; Allore, H.G.; Montgomery, R.R. Dysregulation of Human Toll-like Receptor Function in Aging. Ageing Res. Rev. 2011, 10, 346–353. [Google Scholar] [CrossRef]
  22. Dunston, C.R.; Griffiths, H.R. The Effect of Ageing on Macrophage Toll-like Receptor-Mediated Responses in the Fight against Pathogens. Clin. Exp. Immunol. 2010, 161, 407–416. [Google Scholar] [CrossRef]
  23. Kim, H.-J.; Kim, H.; Lee, J.-H.; Hwangbo, C. Toll-like Receptor 4 (TLR4): New Insight Immune and Aging. Immun. Ageing 2023, 20, 67. [Google Scholar] [CrossRef]
  24. Bailey, K.L.; Smith, L.M.; Heires, A.J.; Katafiasz, D.M.; Romberger, D.J.; LeVan, T.D. Aging Leads to Dysfunctional Innate Immune Responses to TLR2 and TLR4 Agonists. Aging Clin. Exp. Res. 2019, 31, 1185–1193. [Google Scholar] [CrossRef]
  25. Calabria, E.; Mazza, E.M.C.; Dyar, K.A.; Pogliaghi, S.; Bruseghini, P.; Morandi, C.; Salvagno, G.L.; Gelati, M.; Guidi, G.C.; Bicciato, S.; et al. Aging: A Portrait from Gene Expression Profile in Blood Cells. Aging 2016, 8, 1802–1821. [Google Scholar] [CrossRef] [PubMed]
  26. Bickler, S.W.; Cauvi, D.M.; Fisch, K.M.; Prieto, J.M.; Sykes, A.G.; Thangarajah, H.; Lazar, D.A.; Ignacio, R.C.; Gerstmann, D.R.; Ryan, A.F.; et al. Extremes of Age Are Associated with Differences in the Expression of Selected Pattern Recognition Receptor Genes and ACE2, the Receptor for SARS-CoV-2: Implications for the Epidemiology of COVID-19 Disease. BMC Med. Genom. 2021, 14, 138. [Google Scholar] [CrossRef] [PubMed]
  27. Metcalf, T.U.; Wilkinson, P.A.; Cameron, M.J.; Ghneim, K.; Chiang, C.; Wertheimer, A.M.; Hiscott, J.B.; Nikolich-Zugich, J.; Haddad, E.K. Human Monocyte Subsets Are Transcriptionally and Functionally Altered in Aging in Response to Pattern Recognition Receptor Agonists. J. Immunol. 2017, 199, 1405–1417. [Google Scholar] [CrossRef]
  28. Wang, C.; Cheng, Y.; Li, B.; Qiu, X.; Hu, H.; Zhang, X.; Lu, Z.; Zheng, F. Transcriptional Characteristics and Functional Validation of Three Monocyte Subsets during Aging. Immun. Ageing 2023, 20, 50. [Google Scholar] [CrossRef]
  29. Lissner, M.M.; Thomas, B.J.; Wee, K.; Tong, A.-J.; Kollmann, T.R.; Smale, S.T. Age-Related Gene Expression Differences in Monocytes from Human Neonates, Young Adults, and Older Adults. PLoS ONE 2015, 10, e0132061. [Google Scholar] [CrossRef]
  30. Harris, T.B.; Ferrucci, L.; Tracy, R.P.; Corti, M.C.; Wacholder, S.; Ettinger, W.H.; Heimovitz, H.; Cohen, H.J.; Wallace, R. Associations of Elevated Interleukin-6 and C-Reactive Protein Levels with Mortality in the Elderly. Am. J. Med. 1999, 106, 506–512. [Google Scholar] [CrossRef] [PubMed]
  31. Jiménez-Dalmaroni, M.J.; Gerswhin, M.E.; Adamopoulos, I.E. The Critical Role of Toll-like Receptors—From Microbial Recognition to Autoimmunity: A Comprehensive Review. Autoimmun. Rev. 2016, 15, 1–8. [Google Scholar] [CrossRef]
  32. Compté, N.; Zouaoui Boudjeltia, K.; Vanhaeverbeek, M.; De Breucker, S.; Tassignon, J.; Trelcat, A.; Pepersack, T.; Goriely, S. Frailty in Old Age Is Associated with Decreased Interleukin-12/23 Production in Response to Toll-Like Receptor Ligation. PLoS ONE 2013, 8, e65325. [Google Scholar] [CrossRef][Green Version]
  33. Verschoor, C.P.; Johnstone, J.; Millar, J.; Parsons, R.; Lelic, A.; Loeb, M.; Bramson, J.L.; Bowdish, D.M.E. Alterations to the Frequency and Function of Peripheral Blood Monocytes and Associations with Chronic Disease in the Advanced-Age, Frail Elderly. PLoS ONE 2014, 9, e104522. [Google Scholar] [CrossRef]
  34. Reitsema, R.D.; Kumawat, A.K.; Hesselink, B.-C.; van Baarle, D.; van Sleen, Y. Effects of Ageing and Frailty on Circulating Monocyte and Dendritic Cell Subsets. NPJ Aging 2024, 10, 17. [Google Scholar] [CrossRef]
  35. Lukyanova, S.O.; Artemyeva, O.V.; Strazhesko, I.D.; Nasaeva, E.D.; Grechenko, V.V.; Gankovskaya, L.V. Expression of TLR2, IL-1β, and IL-10 Genes as a Possible Factor of Successful or Pathological Aging in Nonagenarians. Bull. Exp. Biol. Med. 2024, 176, 505–508. [Google Scholar] [CrossRef] [PubMed]
  36. Nidadavolu, L.S.; Feger, D.; Chen, D.; Wu, Y.; Grodstein, F.; Gross, A.L.; Bennett, D.A.; Walston, J.D.; Oh, E.S.; Abadir, P.M. Associations between Circulating Cell-Free Mitochondrial DNA, Inflammatory Markers, and Cognitive and Physical Outcomes in Community Dwelling Older Adults. Immun. Ageing 2023, 20, 24. [Google Scholar] [CrossRef] [PubMed]
  37. Picca, A.; Guerra, F.; Calvani, R.; Bucci, C.; Lo Monaco, M.R.; Bentivoglio, A.R.; Coelho-Júnior, H.J.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial Dysfunction and Aging: Insights from the Analysis of Extracellular Vesicles. Int. J. Mol. Sci. 2019, 20, 805. [Google Scholar] [CrossRef]
  38. Gomez, C.R. Role of Heat Shock Proteins in Aging and Chronic Inflammatory Diseases. Geroscience 2021, 43, 2515–2532. [Google Scholar] [CrossRef]
  39. Roh, J.S.; Sohn, D.H. Damage-Associated Molecular Patterns in Inflammatory Diseases. Immune Netw. 2018, 18, e27. [Google Scholar] [CrossRef] [PubMed]
  40. Murao, A.; Aziz, M.; Wang, H.; Brenner, M.; Wang, P. Release Mechanisms of Major DAMPs. Apoptosis 2021, 26, 152–162. [Google Scholar] [CrossRef]
  41. Chen, Z.; Behrendt, R.; Wild, L.; Schlee, M.; Bode, C. Cytosolic Nucleic Acid Sensing as Driver of Critical Illness: Mechanisms and Advances in Therapy. Signal Transduct. Target. Ther. 2025, 10, 90. [Google Scholar] [CrossRef] [PubMed]
  42. Fan, Z.; Yang, J.; Guo, Y.; Liu, Y.; Zhong, X. Altered Levels of Circulating Mitochondrial DNA in Elderly People with Sarcopenia: Association with Mitochondrial Impairment. Exp. Gerontol. 2022, 163, 111802. [Google Scholar] [CrossRef]
  43. De Gaetano, A.; Solodka, K.; Zanini, G.; Selleri, V.; Mattioli, A.V.; Nasi, M.; Pinti, M. Molecular Mechanisms of MtDNA-Mediated Inflammation. Cells 2021, 10, 2898. [Google Scholar] [CrossRef]
  44. Tao, G.; Liao, W.; Hou, J.; Jiang, X.; Deng, X.; Chen, G.; Ding, C. Advances in Crosstalk among Innate Immune Pathways Activated by Mitochondrial DNA. Heliyon 2024, 10, e24029. [Google Scholar] [CrossRef]
  45. Albuquerque-Souza, E.; Crump, K.E.; Rattanaprukskul, K.; Li, Y.; Shelling, B.; Xia-Juan, X.; Jiang, M.; Sahingur, S.E. TLR9 Mediates Periodontal Aging by Fostering Senescence and Inflammaging. J. Dent. Res. 2022, 101, 1628–1636. [Google Scholar] [CrossRef]
  46. Behl, T.; Sharma, E.; Sehgal, A.; Kaur, I.; Kumar, A.; Arora, R.; Pal, G.; Kakkar, M.; Kumar, R.; Bungau, S. Expatiating the Molecular Approaches of HMGB1 in Diabetes Mellitus: Highlighting Signalling Pathways via RAGE and TLRs. Mol. Biol. Rep. 2021, 48, 1869–1881. [Google Scholar] [CrossRef] [PubMed]
  47. Zou, Y.; Wu, Y.; Wei, A.; Nie, H.; Hui, S.; Liu, C.; Deng, T. Serum HMGB1 as a Predictor for Postoperative Delirium in Elderly Patients Undergoing Total Hip Arthroplasty Surgery. Adv. Clin. Exp. Med. 2024, 34, 361–368. [Google Scholar] [CrossRef]
  48. Jia, Y.; Li, D.; Yu, J.; Liu, Y.; Li, F.; Li, W.; Zhang, Q.; Gao, Y.; Zhang, W.; Zeng, Z.; et al. Subclinical Cardiovascular Disease and Frailty Risk: The Atherosclerosis Risk in Communities Study. BMC Geriatr. 2022, 22, 321. [Google Scholar] [CrossRef]
  49. Zheng, J.; Wang, J.; Liu, H.; Chen, F.; Wang, H.; Chen, S.; Xie, J.; Zheng, Z.; Li, Z. Alarmins S100A8/A9 Promote Intervertebral Disc Degeneration and Inflammation-Related Pain in a Rat Model through Toll-like Receptor-4 and Activation of the NF-ΚB Signaling Pathway. Osteoarthr. Cartil. 2022, 30, 998–1011. [Google Scholar] [CrossRef] [PubMed]
  50. Gupta, S.; Tsoporis, J.N.; Jia, S.-H.; dos Santos, C.C.; Parker, T.G.; Marshall, J.C. Toll-Like Receptors, Associated Biochemical Signaling Networks, and S100 Ligands. Shock 2021, 56, 167–177. [Google Scholar] [CrossRef]
  51. Bockstiegel, J.; Engelhardt, J.; Weindl, G. P2X7 Receptor Activation Leads to NLRP3-Independent IL-1β Release by Human Macrophages. Cell Commun. Signal. 2023, 21, 335. [Google Scholar] [CrossRef]
  52. Fisch, D.; Zhang, T.; Sun, H.; Ma, W.; Tan, Y.; Gygi, S.P.; Higgins, D.E.; Kagan, J.C. Molecular Definition of the Endogenous Toll-like Receptor Signalling Pathways. Nature 2024, 631, 635–644. [Google Scholar] [CrossRef]
  53. Mushenkova, N.V.; Bezsonov, E.E.; Orekhova, V.A.; Popkova, T.V.; Starodubova, A.V.; Orekhov, A.N. Recognition of Oxidized Lipids by Macrophages and Its Role in Atherosclerosis Development. Biomedicines 2021, 9, 915. [Google Scholar] [CrossRef]
  54. Wicherska-Pawłowska, K.; Wróbel, T.; Rybka, J. Toll-Like Receptors (TLRs), NOD-Like Receptors (NLRs), and RIG-I-Like Receptors (RLRs) in Innate Immunity. TLRs, NLRs, and RLRs Ligands as Immunotherapeutic Agents for Hematopoietic Diseases. Int. J. Mol. Sci. 2021, 22, 13397. [Google Scholar] [CrossRef]
  55. Salminen, A.; Kauppinen, A.; Kaarniranta, K. Emerging Role of NF-ΚB Signaling in the Induction of Senescence-Associated Secretory Phenotype (SASP). Cell Signal 2012, 24, 835–845. [Google Scholar] [CrossRef]
  56. Heo, Y.J.; Lee, N.; Choi, S.-E.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Amphiregulin Induces INOS and COX-2 Expression through NF-ΚB and MAPK Signaling in Hepatic Inflammation. Mediators Inflamm. 2023, 2023, 2364121. [Google Scholar] [CrossRef]
  57. Perkins, D.J.; Richard, K.; Hansen, A.-M.; Lai, W.; Nallar, S.; Koller, B.; Vogel, S.N. Autocrine–Paracrine Prostaglandin E2 Signaling Restricts TLR4 Internalization and TRIF Signaling. Nat. Immunol. 2018, 19, 1309–1318. [Google Scholar] [CrossRef]
  58. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed]
  59. Bleve, A.; Motta, F.; Durante, B.; Pandolfo, C.; Selmi, C.; Sica, A. Immunosenescence, Inflammaging, and Frailty: Role of Myeloid Cells in Age-Related Diseases. Clin. Rev. Allergy Immunol. 2022, 64, 123–144. [Google Scholar] [CrossRef] [PubMed]
  60. Fang, W.; Tseng, Y.; Lee, T.; Fu, Y.; Chang, W.; Lo, W.; Lin, C.; Lo, Y. Triptolide Prevents LPS-induced Skeletal Muscle Atrophy via Inhibiting NF-κB/TNF-α and Regulating Protein Synthesis/Degradation Pathway. Br. J. Pharmacol. 2021, 178, 2998–3016. [Google Scholar] [CrossRef] [PubMed]
  61. Dongre, U.J. Adipokines in Insulin Resistance: Current Updates. Biosci. Biotechnol. Res. Asia 2021, 18, 357–366. [Google Scholar] [CrossRef]
  62. Fischer, R.; Maier, O. Interrelation of Oxidative Stress and Inflammation in Neurodegenerative Disease: Role of TNF. Oxid. Med. Cell Longev. 2015, 2015, 610813. [Google Scholar] [CrossRef]
  63. van Sleen, Y.; Shetty, S.A.; van der Heiden, M.; Venema, M.C.A.; Gutiérrez-Melo, N.; Toonen, E.J.M.; van Beek, J.; Buisman, A.-M.; van Baarle, D.; Sauce, D. Frailty Is Related to Serum Inflammageing Markers: Results from the VITAL Study. Immun. Ageing 2023, 20, 68. [Google Scholar] [CrossRef]
  64. Paudel, Y.N.; Angelopoulou, E.; Piperi, C.; Othman, I.; Aamir, K.; Shaikh, M.F. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s Disease (AD): From Risk Factors to Therapeutic Targeting. Cells 2020, 9, 383. [Google Scholar] [CrossRef]
  65. Rai, V.; Agrawal, D.K. The Role of Damage- and Pathogen-Associated Molecular Patterns in Inflammation-Mediated Vulnerability of Atherosclerotic Plaques. Can. J. Physiol. Pharmacol. 2017, 95, 1245–1253. [Google Scholar] [CrossRef]
  66. Meftahi, G.H.; Jangravi, Z.; Sahraei, H.; Bahari, Z. The Possible Pathophysiology Mechanism of Cytokine Storm in Elderly Adults with COVID-19 Infection: The Contribution of “Inflame-Aging”. Inflamm. Res. 2020, 69, 825–839. [Google Scholar] [CrossRef]
  67. Panyod, S.; Wu, W.-K.; Hsieh, Y.-C.; Tseng, Y.-J.; Peng, S.-Y.; Chen, R.-A.; Huang, H.-S.; Chen, Y.-H.; Shen, T.-C.D.; Ho, C.-T.; et al. Ginger Essential Oil Prevents NASH Progression by Blocking the NLRP3 Inflammasome and Remodeling the Gut Microbiota-LPS-TLR4 Pathway in Mice. Nutr. Diabetes 2024, 14, 65. [Google Scholar] [CrossRef] [PubMed]
  68. Shi, X.-Y.; Zheng, X.-M.; Liu, H.-J.; Han, X.; Zhang, L.; Hu, B.; Li, S. Rotundic Acid Improves Nonalcoholic Steatohepatitis in Mice by Regulating Glycolysis and the TLR4/AP1 Signaling Pathway. Lipids Health Dis. 2023, 22, 214. [Google Scholar] [CrossRef]
  69. Liu, Y.; Liu, N.; Liu, Y.; He, H.; Luo, Z.; Liu, W.; Song, N.; Ju, M. Ginsenoside Rb1 Reduces D-GalN/LPS-Induced Acute Liver Injury by Regulating TLR4/NF-ΚB Signaling and NLRP3 Inflammasome. J. Clin. Transl. Hepatol. 2022, 10, 474–485. [Google Scholar] [CrossRef] [PubMed]
  70. Zhu, H.; Bian, C.; Yuan, J.; Chu, W.; Xiang, X.; Chen, F.; Wang, C.; Feng, H.; Lin, J. Curcumin Attenuates Acute Inflammatory Injury by Inhibiting the TLR4/MyD88/NF-ΚB Signaling Pathway in Experimental Traumatic Brain Injury. J. Neuroinflamm. 2014, 11, 59. [Google Scholar] [CrossRef] [PubMed]
  71. WeiGang, H.; KaiQiang, L.; XueYou, H.; JiaHan, X.; TaiXin, Z.; YingKai, D.; JunYi, H.; MoYan, J.; JiaChen, W.; Xin, W.; et al. Therapeutic Potential of a Triazole Curcumin in Inflammation: Decreased LPS-Induced Acute Lung Injury in Mice by Targeting MD2/TLR4. Arab. J. Chem. 2023, 16, 105076. [Google Scholar] [CrossRef]
  72. Tu, C.; Han, B.; Yao, Q.; Zhang, Y.; Liu, H.; Zhang, S. Curcumin Attenuates Concanavalin A-Induced Liver Injury in Mice by Inhibition of Toll-like Receptor (TLR) 2, TLR4 and TLR9 Expression. Int. Immunopharmacol. 2012, 12, 151–157. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, J.; Yuan, Y.; Gong, X.; Zhang, L.; Zhou, Q.; Wu, S.; Zhang, X.; Hu, J.; Kuang, G.; Yin, X.; et al. Baicalin and Its Nanoliposomes Ameliorates Nonalcoholic Fatty Liver Disease via Suppression of TLR4 Signaling Cascade in Mice. Int. Immunopharmacol. 2020, 80, 106208. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, T.; Zhu, Q.; Shao, Y.; Wang, K.; Wu, Y. Paeoniflorin Prevents TLR2/4-Mediated Inflammation in Type 2 Diabetic Nephropathy. Biosci. Trends 2017, 11, 308–318. [Google Scholar] [CrossRef] [PubMed]
  75. Xie, T.; Li, K.; Gong, X.; Jiang, R.; Huang, W.; Chen, X.; Tie, H.; Zhou, Q.; Wu, S.; Wan, J.; et al. Paeoniflorin Protects against Liver Ischemia/Reperfusion Injury in Mice via Inhibiting HMGB1-TLR4 Signaling Pathway. Phytother. Res. 2018, 32, 2247–2255. [Google Scholar] [CrossRef]
  76. Ma, J.-Q.; Li, Z.; Xie, W.-R.; Liu, C.-M.; Liu, S.-S. Quercetin Protects Mouse Liver against CCl4-Induced Inflammation by the TLR2/4 and MAPK/NF-ΚB Pathway. Int. Immunopharmacol. 2015, 28, 531–539. [Google Scholar] [CrossRef]
  77. Chen, H.; Liu, Q.; Liu, X.; Jin, J. Berberine Attenuates Septic Cardiomyopathy by Inhibiting TLR4/NF-ΚB Signalling in Rats. Pharm. Biol. 2021, 59, 119–126. [Google Scholar] [CrossRef]
  78. Al-dossari, M.H.; Fadda, L.M.; Attia, H.A.; Hasan, I.H.; Mahmoud, A.M. Curcumin and Selenium Prevent Lipopolysaccharide/Diclofenac-Induced Liver Injury by Suppressing Inflammation and Oxidative Stress. Biol. Trace Elem. Res. 2020, 196, 173–183. [Google Scholar] [CrossRef]
  79. Tu, C.; Yao, Q.; Xu, B.; Wang, J.; Zhou, C.; Zhang, S. Protective Effects of Curcumin against Hepatic Fibrosis Induced by Carbon Tetrachloride: Modulation of High-Mobility Group Box 1, Toll-like Receptor 4 and 2 Expression. Food Chem. Toxicol. 2012, 50, 3343–3351. [Google Scholar] [CrossRef] [PubMed]
  80. Li, X.; Liu, H.; Yao, Q.; Xu, B.; Zhang, S.; Tu, C. Quercetin Protects Mice from ConA-Induced Hepatitis by Inhibiting HMGB1-TLR Expression and Down-Regulating the Nuclear Factor Kappa B Pathway. Inflammation 2016, 39, 96–106. [Google Scholar] [CrossRef]
  81. Lee, S.A.; Lee, S.H.; Kim, J.Y.; Lee, W.S. Effects of Glycyrrhizin on Lipopolysaccharide-Induced Acute Lung Injury in a Mouse Model. J. Thorac. Dis. 2019, 11, 1287–1302. [Google Scholar] [CrossRef] [PubMed]
  82. ZHANG, Z.; CHEN, N.; LIU, J.-B.; WU, J.-B.; ZHANG, J.; ZHANG, Y.; JIANG, X. Protective Effect of Resveratrol against Acute Lung Injury Induced by Lipopolysaccharide via Inhibiting the Myd88-Dependent Toll-like Receptor 4 Signaling Pathway. Mol. Med. Rep. 2014, 10, 101–106. [Google Scholar] [CrossRef]
  83. Jingyan, L.; Yujuan, G.; Yiming, Y.; Lingpeng, Z.; Tianhua, Y.; Mingxing, M. Salidroside Attenuates LPS-Induced Acute Lung Injury in Rats. Inflammation 2017, 40, 1520–1531. [Google Scholar] [CrossRef]
  84. Wang, N.; Geng, C.; Sun, H.; Wang, X.; Li, F.; Liu, X. Hesperetin Ameliorates Lipopolysaccharide-Induced Acute Lung Injury in Mice through Regulating the TLR4–MyD88–NF-ΚB Signaling Pathway. Arch. Pharm. Res. 2019, 42, 1063–1070. [Google Scholar] [CrossRef]
  85. Yuan, X.; Zhu, J.; Kang, Q.; He, X.; Guo, D. Protective Effect of Hesperidin Against Sepsis-Induced Lung Injury by Inducing the Heat-Stable Protein 70 (Hsp70)/Toll-Like Receptor 4 (TLR4)/Myeloid Differentiation Primary Response 88 (MyD88) Pathway. Med. Sci. Monit. 2019, 25, 107–114. [Google Scholar] [CrossRef] [PubMed]
  86. Shen, N.; Cheng, A.; Qiu, M.; Zang, G. Allicin Improves Lung Injury Induced by Sepsis via Regulation of the Toll-Like Receptor 4 (TLR4)/Myeloid Differentiation Primary Response 88 (MYD88)/Nuclear Factor Kappa B (NF-ΚB) Pathway. Med. Sci. Monit. 2019, 25, 2567–2576. [Google Scholar] [CrossRef]
  87. Panyod, S.; Wu, W.-K.; Lu, K.-H.; Liu, C.-T.; Chu, Y.-L.; Ho, C.-T.; Hsiao, W.-L.W.; Lai, Y.-S.; Chen, W.-C.; Lin, Y.-E.; et al. Allicin Modifies the Composition and Function of the Gut Microbiota in Alcoholic Hepatic Steatosis Mice. J. Agric. Food Chem. 2020, 68, 3088–3098. [Google Scholar] [CrossRef]
  88. Guo, S.; Jiang, K.; Wu, H.; Yang, C.; Yang, Y.; Yang, J.; Zhao, G.; Deng, G. Magnoflorine Ameliorates Lipopolysaccharide-Induced Acute Lung Injury via Suppressing NF-ΚB and MAPK Activation. Front. Pharmacol. 2018, 9, 982. [Google Scholar] [CrossRef] [PubMed]
  89. Yunhe, F.; Bo, L.; Xiaosheng, F.; Fengyang, L.; Dejie, L.; Zhicheng, L.; Depeng, L.; Yongguo, C.; Xichen, Z.; Naisheng, Z.; et al. The Effect of Magnolol on the Toll-like Receptor 4/Nuclear Factor Kappa B Signaling Pathway in Lipopolysaccharide-Induced Acute Lung Injury in Mice. Eur. J. Pharmacol. 2012, 689, 255–261. [Google Scholar] [CrossRef]
  90. Mohammed, E.T.; Safwat, G.M. Grape Seed Proanthocyanidin Extract Mitigates Titanium Dioxide Nanoparticle (TiO2-NPs)–Induced Hepatotoxicity Through TLR-4/NF-ΚB Signaling Pathway. Biol. Trace Elem. Res. 2020, 196, 579–589. [Google Scholar] [CrossRef]
  91. Ye, H.-Y.; Jin, J.; Jin, L.-W.; Chen, Y.; Zhou, Z.-H.; Li, Z.-Y. Chlorogenic Acid Attenuates Lipopolysaccharide-Induced Acute Kidney Injury by Inhibiting TLR4/NF-ΚB Signal Pathway. Inflammation 2017, 40, 523–529. [Google Scholar] [CrossRef]
  92. Fan, H.-Y.; Qi, D.; Yu, C.; Zhao, F.; Liu, T.; Zhang, Z.-K.; Yang, M.-Y.; Zhang, L.-M.; Chen, D.-Q.; Du, Y. Paeonol Protects Endotoxin-Induced Acute Kidney Injury: Potential Mechanism of Inhibiting TLR4-NF-κB Signal Pathway. Oncotarget 2016, 7, 39497–39510. [Google Scholar] [CrossRef]
  93. Cui, H.-X.; Chen, J.-H.; Li, J.-W.; Cheng, F.-R.; Yuan, K. Protection of Anthocyanin from Myrica Rubra against Cerebral Ischemia-Reperfusion Injury via Modulation of the TLR4/NF-ΚB and NLRP3 Pathways. Molecules 2018, 23, 1788. [Google Scholar] [CrossRef]
  94. Yubolphan, R.; Kobroob, A.; Kongkaew, A.; Chiranthanut, N.; Jinadang, N.; Wongmekiat, O. Berberine Mitigates Sepsis-Associated Acute Kidney Injury in Aged Rats by Preserving Mitochondrial Integrity and Inhibiting TLR4/NF-ΚB and NLRP3 Inflammasome Activations. Antioxidants 2024, 13, 1398. [Google Scholar] [CrossRef]
  95. Wang, X.-P.; Lei, F.; Du, F.; Chai, Y.-S.; Jiang, J.-F.; Wang, Y.-G.; Yu, X.; Yan, X.-J.; Xing, D.-M.; Du, L.-J. Protection of Gastrointestinal Mucosa from Acute Heavy Alcohol Consumption: The Effect of Berberine and Its Correlation with TLR2, 4/IL1β-TNFα Signaling. PLoS ONE 2015, 10, e0134044. [Google Scholar] [CrossRef]
  96. Kim, T.-H.; Yoon, S.-J.; Lee, S.-M. Genipin Attenuates Sepsis by Inhibiting Toll-Like Receptor Signaling. Mol. Med. 2012, 18, 455–465. [Google Scholar] [CrossRef]
  97. Zhao, X.; Yin, L.; Fang, L.; Xu, L.; Sun, P.; Xu, M.; Liu, K.; Peng, J. Protective Effects of Dioscin against Systemic Inflammatory Response Syndromevia Adjusting TLR2/MyD88/NF-κb Signal Pathway. Int. Immunopharmacol. 2018, 65, 458–469. [Google Scholar] [CrossRef] [PubMed]
  98. Lei, J.; Xie, Q.; Li, Q.; Qin, S.; Wu, J.; Jiang, H.; Yu, B.; Luo, W. Resveratrol Alleviates Liver Fibrosis by Targeting Cross-Talk Between TLR2/MyD88/ERK and NF-κB/NLRP3 Inflammasome Pathways in Macrophages. J. Biochem. Mol. Toxicol. 2025, 39, e70208. [Google Scholar] [CrossRef]
  99. Fei, L.; Jifeng, F.; Tiantian, W.; Yi, H.; Linghui, P. Glycyrrhizin Ameliorate Ischemia Reperfusion Lung Injury through Downregulate TLR2 Signaling Cascade in Alveolar Macrophages. Front. Pharmacol. 2017, 8, 389. [Google Scholar] [CrossRef] [PubMed]
  100. Zhang, J.; Sheng, Y.; Shi, L.; Zheng, Z.; Chen, M.; Lu, B.; Ji, L. Quercetin and Baicalein Suppress Monocrotaline-Induced Hepatic Sinusoidal Obstruction Syndrome in Rats. Eur. J. Pharmacol. 2017, 795, 160–168. [Google Scholar] [CrossRef] [PubMed]
  101. Li, S.; Feng, G.; Zhang, M.; Zhang, X.; Lu, J.; Feng, C.; Zhu, F. Oxymatrine Attenuates TNBS-Induced Colinutis in Rats through TLR9/Myd88/NF-ΚB Signal Pathway. Hum. Exp. Toxicol. 2022, 41, 9603271221078866. [Google Scholar] [CrossRef] [PubMed]
  102. Li, H.; Sun, H.; Xu, Y.; Xing, G.; Wang, X. Curcumin Plays a Protective Role against Septic Acute Kidney Injury by Regulating the TLR9 Signaling Pathway. Transl. Androl. Urol. 2021, 10, 2103–2112. [Google Scholar] [CrossRef]
  103. Gu, J.; Ran, X.; Deng, J.; Zhang, A.; Peng, G.; Du, J.; Wen, D.; Jiang, B.; Xia, F. Glycyrrhizin Alleviates Sepsis-Induced Acute Respiratory Distress Syndrome via Suppressing of HMGB1/TLR9 Pathways and Neutrophils Extracellular Traps Formation. Int. Immunopharmacol. 2022, 108, 108730. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, W.; Wu, C.; Wang, Q.; Kuang, L.; Le, A. Tanshinone IIA Relieves Arthritis by Inhibiting Autophagy of Fibroblast-like Synoviocytes via Matrix Metalloproteinase9/Receptor for Advanced Glycation End Product/Toll-like Receptor 9 Signal Axis in Mice with Collagen-induced Arthritis. Phytother. Res. 2023, 37, 1391–1404. [Google Scholar] [CrossRef]
  105. Iskender, H.; Dokumacioglu, E.; Terim Kapakin, K.A.; Yenice, G.; Mohtare, B.; Bolat, I.; Hayirli, A. Effects of Oleanolic Acid on Inflammation and Metabolism in Diabetic Rats. Biotech. Histochem. 2022, 97, 269–276. [Google Scholar] [CrossRef]
  106. Zhang, M.; Zhang, Y.; Peng, J.; Huang, Y.; Gong, Z.; Lu, H.; Han, L.; Wang, D. Gastrodin against Oxidative Stress-Inflammation Crosstalk via Inhibiting MtDNA/TLR9 and JAK2/STAT3 Signaling to Ameliorate Ischemic Stroke Injury. Int. Immunopharmacol. 2024, 141, 113012. [Google Scholar] [CrossRef]
  107. Dokumacioğlu, E.; İskender, H.; Terim Kapakin, K.A.; Yenice, G.; Mokthare, B.; Bolat, İ.; Hayirli, A. Effect of Betulinic Acid Administration on TLR-9/NF-ΚB/IL-18 Levels in Experimental Liver Injury. Turk. J. Med. Sci. 2021, 51, 1543–1552. [Google Scholar] [CrossRef]
  108. PubChem. Available online: https://pubchem.ncbi.nlm.nih.gov/ (accessed on 12 November 2025).
  109. Brubaker, D.K.; Lauffenburger, D.A. Translating Preclinical Models to Humans. Science 2020, 367, 742–743. [Google Scholar] [CrossRef]
  110. Doherty, T.M.; Weinberger, B.; Didierlaurent, A.; Lambert, P.-H. Age-Related Changes in the Immune System and Challenges for the Development of Age-Specific Vaccines. Ann. Med. 2025, 57, 2477300. [Google Scholar] [CrossRef] [PubMed]
  111. Kaur, A.; Baldwin, J.; Brar, D.; Salunke, D.B.; Petrovsky, N. Toll-like Receptor (TLR) Agonists as a Driving Force behind next-Generation Vaccine Adjuvants and Cancer Therapeutics. Curr. Opin. Chem. Biol. 2022, 70, 102172. [Google Scholar] [CrossRef]
  112. Kuzmich, N.; Sivak, K.; Chubarev, V.; Porozov, Y.; Savateeva-Lyubimova, T.; Peri, F. TLR4 Signaling Pathway Modulators as Potential Therapeutics in Inflammation and Sepsis. Vaccines 2017, 5, 34. [Google Scholar] [CrossRef]
  113. Shi, H.; Dong, L.; Dang, X.; Liu, Y.; Jiang, J.; Wang, Y.; Lu, X.; Guo, X. Effect of Chlorogenic Acid on LPS-Induced Proinflammatory Signaling in Hepatic Stellate Cells. Inflamm. Res. 2013, 62, 581–587. [Google Scholar] [CrossRef] [PubMed]
  114. Fu, Y.; Zhou, E.; Wei, Z.; Song, X.; Liu, Z.; Wang, T.; Wang, W.; Zhang, N.; Liu, G.; Yang, Z. Glycyrrhizin Inhibits Lipopolysaccharide-Induced Inflammatory Response by Reducing TLR4 Recruitment into Lipid Rafts in RAW264.7 Cells. Biochim. Et Biophys. Acta (BBA)—Gen. Subj. 2014, 1840, 1755–1764. [Google Scholar] [CrossRef] [PubMed]
  115. Federico, S.; Pozzetti, L.; Papa, A.; Carullo, G.; Gemma, S.; Butini, S.; Campiani, G.; Relitti, N. Modulation of the Innate Immune Response by Targeting Toll-like Receptors: A Perspective on Their Agonists and Antagonists. J. Med. Chem. 2020, 63, 13466–13513. [Google Scholar] [CrossRef] [PubMed]
  116. Gong, J.; Li, J.; Dong, H.; Chen, G.; Qin, X.; Hu, M.; Yuan, F.; Fang, K.; Wang, D.; Jiang, S.; et al. Inhibitory Effects of Berberine on Proinflammatory M1 Macrophage Polarization through Interfering with the Interaction between TLR4 and MyD88. BMC Complement. Altern. Med. 2019, 19, 314. [Google Scholar] [CrossRef]
  117. Wang, L.; Li, N.; Lin, D.; Zang, Y. Curcumin Protects against Hepatic Ischemia/Reperfusion Induced Injury through Inhibiting TLR4/NF-ΚB Pathway. Oncotarget 2017, 8, 65414–65420. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of TLR2, TLR4, and TLR9 signaling pathways. Legend: DAMPs, Damage-associated molecular patterns; HMGBl, High mobility group box 1; HSPs, Heat shock proteins; IFN, interferon; IL, interleukin; MAPK, Mitogen-activated protein kinase; mtDNA, Mitochondrial DNA; MyD88, Myeloid differentiation primary response 88; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; PAMPs, Pathogen-associated molecular patterns; S100A8/A9, S100 calcium-binding proteins A8 and A9; TLR, Toll-like receptor; TRIF, TIR-domain-containing adapter-inducing interferon-β; TNF-α, Tumor necrosis factor alpha.
Figure 1. Schematic representation of TLR2, TLR4, and TLR9 signaling pathways. Legend: DAMPs, Damage-associated molecular patterns; HMGBl, High mobility group box 1; HSPs, Heat shock proteins; IFN, interferon; IL, interleukin; MAPK, Mitogen-activated protein kinase; mtDNA, Mitochondrial DNA; MyD88, Myeloid differentiation primary response 88; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; PAMPs, Pathogen-associated molecular patterns; S100A8/A9, S100 calcium-binding proteins A8 and A9; TLR, Toll-like receptor; TRIF, TIR-domain-containing adapter-inducing interferon-β; TNF-α, Tumor necrosis factor alpha.
Ijms 26 11305 g001
Figure 2. Mechanisms by which flavonoids modulate TLR2/4/9 signaling to reduce chronic inflammation. Legend: DAMPs, Damage-Associated Molecular Patterns; HMGB1, High-Mobility Group Box 1; IL-1β, Interleukin-1 beta; IL-6, Interleukin-6; IL-10, Interleukin-10; MAPK, Mitogen-Activated Protein Kinase; MyD88, Myeloid Differentiation Primary Response 88; NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells; TLR, Toll-Like Receptor; TNF-α, Tumor Necrosis Factor alpha.
Figure 2. Mechanisms by which flavonoids modulate TLR2/4/9 signaling to reduce chronic inflammation. Legend: DAMPs, Damage-Associated Molecular Patterns; HMGB1, High-Mobility Group Box 1; IL-1β, Interleukin-1 beta; IL-6, Interleukin-6; IL-10, Interleukin-10; MAPK, Mitogen-Activated Protein Kinase; MyD88, Myeloid Differentiation Primary Response 88; NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells; TLR, Toll-Like Receptor; TNF-α, Tumor Necrosis Factor alpha.
Ijms 26 11305 g002
Table 1. Age-associated alterations in Toll-like receptor (TLR) expression and signaling across different human cell types and tissues.
Table 1. Age-associated alterations in Toll-like receptor (TLR) expression and signaling across different human cell types and tissues.
Body Site/Cell Type (Human)TLRDirection with AgeReference
Peripheral blood—myeloid DCsTLR1, TLR2, TLR3, TLR8↓ expression and function[10]
Peripheral blood—plasmacytoid DCsTLR9 (and TLR7 responsiveness)↓ responsiveness/expression signals[11]
Peripheral blood—NeutrophilsTLR1↓ surface expression[12]
Peripheral blood—MonocytesTLR5↑ expression and signaling[13]
Skeletal muscle (vastus lateralis)TLR4↑ expression/signaling with aging[14,15]
Legend: ↓, decreased; ↑, increased; DCs, Dendritic cells; TLR, Toll-like receptor.
Table 2. Summary of human omics studies assessing age-related differences in Toll-like receptor (TLR) expression and signaling.
Table 2. Summary of human omics studies assessing age-related differences in Toll-like receptor (TLR) expression and signaling.
Author (Year)Sample and PopulationOmics MethodTLR-Relevant Findings
Calabria et al. (2016) [25]Whole blood
Healthy adults (46 ± 3 y, N = 11) vs. healthy elderly (68 ± 4 y, N = 9);
Whole-blood microarray + GSEA + qPCREnrichment of the Toll pathway gene set with higher expression of TLR4, TLR6, TLR9, MYD88, IKBKG (NEMO), and NFKBIA in the elderly.
Bickler et al. (2021) [26]Human dermal fibroblast lines spanning ≤ 10 y (N = 14) to ≥ 80 y (N = 33) GSE113957)Bulk RNA-seq reanalysis≥80 y vs. ≤10 y groups:
Significantly upregulated: TLR3, TLR4, IFIH1
TLR4 showed the strongest effect.
Negatively correlated: NOD1, cGAS
Metcalf et al. (2017) [27]Sorted monocyte subsets
Healthy non-frail adults (21–40 y) vs. older adults (≥65 y);
Global transcriptomics after PRR agonists (TLR4, TLR7/8, RIG-I)Surface TLR expression (TLR3, TLR4, TLR7) did not vary with age.
Old vs. young:
↓ TLR-induced cytokine response (IL-1β, IL-6, IFN-γ, TNF-α)
↓ TLR-associated antiviral/interferon genes
↓ NF-κB target genes
↑ Oxidative stress–related transcripts
Wang et al. (2023) [28]Three monocyte subsets; CD14+ monocytes (N = 1202, 44–83 y)
Healthy young (19–30 y) vs. elderly (55–86 y);
Bulk transcriptomics + pathway analysisOld monocytes vs. young monocytes:
↓ IFN-β, IL-1β and IFN-γ response to viral exposure, while ↑ IL-6.
Legend: ↓, decreased; ↑, increased; cGAS, cyclic GMP-AMP synthase; CD14+, cluster of differentiation 14 positive; GSEA, gene set enrichment analysis; IFIH1, interferon induced with helicase C domain 1 (MDA5); IFN-β, interferon-beta; IFN-γ, interferon-gamma; IKBKG (NEMO), inhibitor of nuclear factor kappa-B kinase subunit gamma (NF-κB essential modulator); IL-1β, interleukin-1 beta; IL-6, interleukin-6; MYD88, myeloid differentiation primary response 88; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NFKBIA, nuclear factor kappa-B inhibitor alpha; NOD1, nucleotide-binding oligomerization domain-containing protein 1; PRR, pattern recognition receptor; qPCR, quantitative polymerase chain reaction; RIG-I, retinoic acid–inducible gene I (DDX58); RNA-seq, RNA sequencing; TLR, Toll-like receptor; TNF-α, tumor necrosis factor-alpha.
Table 3. Observational studies examining TLR expression, inflammatory responses, and frailty in older adults.
Table 3. Observational studies examining TLR expression, inflammatory responses, and frailty in older adults.
Study (Year)SettingsDesign and Frailty MeasureTLR TargetMain Finding Related to Frailty
Compté et al., 2013 [32]Belgium; 100 participants aged 23–96 years
Subset analysis: 52 participants >75 years old (27 non-frail, 25 frail).
Cross-sectional observational study.
Frailty assessed by ISAR scale.
Whole-blood ex vivo; stimulation with LPS (TLR4 ligand) + R848 (TLR7/8 ligand).Elderly vs. young:
TLR4, NF-κB, and p38 expression, and Erk phosphorylation remain stable on monocytes and cDCs
↓ IL-12p70, IL-23 (even more in frail elderly)
Verschoor et al., 2014 [33]Canada; 129 participants “advanced-age, frail elderly” (61–100 y) vs. young adults (19–59 y)Cross-sectional observational study.
Frailty assessed using the Clinical Frailty Scale (score ≥ 4)
PBMCs—monocytes and DCs;
stimulation with Pam3CSK4 (TLR2 agonist) and LPS (TLR4 agonist).
Elderly vs. young:
↑ TLR2 on myeloid DCs
↑ TLR4 on classical monocytes
↓ CCR2 (no change between seniors ↔ frail)
↓ CX3CR1 only in frail elderly
Reitsema et al., 2024 [34]Netherlands and Sweden; 45 participants
Healthy young controls (median age 29 y) vs. healthy (73 y) or frail (76 y) older adults
Observational cross-sectional study.
Frailty assessed using the Tilburg Frailty Indicator; Groningen Frailty Indicator, and Fried Frailty Phenotype.
PBMCs—monocyte subsetsElderly vs. young:
↑ TLR2 on classical monocytes and cDC2
TLR4 ↔ (no significant change)
Frailty vs. healthy old age:
No significant differences in subset frequencies or other markers (TLR2, TLR4, CD86, CD11c, HLA-DR, PD-L1).
Elderly vs. young:
Lukyanova et al. (2024) [35]Russia
219 nonagenarians (mean age = 92.1 y); control 24 healthy young donors (mean age = 22.5 y).
Observational cross-sectional study
nonagenarians categorized as with/without frailty and as successful vs. pathological aging based on presence of four geriatric syndromes (frailty, dementia, sarcopenia, reduced functional activity).
TLR2 gene expression assessed in peripheral blood leukocytes.↑ IL1β and TLR2 expression
↓ IL10 expression
Frailty vs. non-frailty:
↑ IL1β and TLR2 expression
Successful vs. pathological aging:
↑ IL10 expression
Similar IL1β and TLR2 levels of expression
Nidadavolu et al. (2023) [36]USA; community-dwelling older adults (N = 672; mean age 80.4 ± 7.2 y) Observational, cross-sectional and longitudinal analyses.
Frailty measured as a composite z-score including grip strength, timed walk, body mass index, and fatigue
Indirect innate immune activation assessed via circulating cell-free mtDNA Elevated mtDNA levels correlated with ↑ frailty, ↓ gait speed, ↓ grip strength, ↓ cognitive performance, and ↑ mortality risk.
cf-mtDNA-induced activation of TLR9:
↑ CRP, ↑ TNF-α, and ↑ IL-6
Legend: ↓, decreased; ↑, increased; ↔, unchanged; ACE2, angiotensin-converting enzyme 2; CCR2, C-C chemokine receptor type 2; cDC, conventional dendritic cell; cDC2, conventional dendritic cell type 2; cf-mtDNA, circulating cell-free mitochondrial DNA; CRP, C-reactive protein; CX3CR1, C-X3-C motif chemokine receptor 1; DC, dendritic cell; Erk, extracellular signal-regulated kinase; HLA-DR, human leukocyte antigen—DR isotype; IL-1β, interleukin-1 beta; IL-6, interleukin-6; IL-10, interleukin-10; IL-12p70, interleukin-12 p70 subunit; IL-23, interleukin-23; ISAR, Identification of Seniors at Risk; LPS, lipopolysaccharide; mtDNA, mitochondrial DNA; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PBMC, peripheral blood mononuclear cell; PD-L1, programmed death-ligand 1; p38, p38 mitogen-activated protein kinase; R848, resiquimod; TLR, Toll-like receptor; TNF-α, tumor necrosis factor-alpha.
Table 4. Natural compounds targeting TLR4 signaling pathways in experimental models of inflammation and tissue injury.
Table 4. Natural compounds targeting TLR4 signaling pathways in experimental models of inflammation and tissue injury.
Natural CompoundSourcesExperimental ModelMode of AdministrationTLR4-Related MechanismMain OutcomesReference
BaicalinScutellaria baicalensis GeorgiMCD-induced NAFLD
C57BL/6J mice
50 mg/kg, orally,
4 weeks
TLR4/MyD88/NF-κB/p38 MAPK inhibition↓ TNF-α
↓ IL-1β
↓ IL-6
↓ CCL2
↓ CXCL2
↓ ICAM
↓ VCAM
↓ ELAM
[73]
Ginger essential oilZingiber officinaleP-HFD/LPS-induced NASH
C57BL/6 J mice
12.5–125 mg/kg,
orally,
12 weeks
TLR4/NF-κB inhibition↓ TNF-α
↓ IL-1β
↓ IL-6
[67]
PaeoniflorinPaeonia lactiflora PallHepatic ischemia/reperfusion injury
C57BL/6 mice
100 mg/kg, orally,
3 times (every 8 h) before surgery
TLR4/HMGB1/ERK1/2/JNK1/2/p38/NF-κB inhibition↓ TNF-α
↓ IL-1β
[75]
Type 2 diabetes-induced nephropathy
db/db mice
15–60 mg,
i.p.,
2 weeks
TLR4/MyD88/IRAK1/NF-κB/iNOS inhibition↓ TNF-α
↓ IL-1β
↓ MCP-1
↓ CD68+
[74]
Rotundic acidIlex rotundaHFD-induced NASH
C57BL/6 mice
30 mg/kg,
orally,
3 weeks
TLR4/MyD88/MAP3K8/MAPK3/AP-1 (c-Fos/c-Jun) inhibition-[68]
Ginsenoside Rb1Panax ginsengD-GalN/LPS-induced acute liver injury
C57BL/6 mice
30–60 mg/kg,
i.p.
3 days before
D-GalN/LPS treatment
TLR4/MyD88/NF-κB/NLRP3 inhibition↓ TNF-α
↓ IL-6
↓ IL-1β
↓ IL-18
[69]
CurcuminCurcuma longa LinnConcanavalin A–induced autoimmune hepatitis
BALB/c mice
200 mg/kg,
orally,
40 min before Concanavalin A injection
TLR4 inhibition↓ TNF-α
↓ IFN-γ
↑ IL-10
↓ Kupffer cell infiltration (F4/80+)
[72]
LPS/DCL-induced hepatotoxicity
Wistar rats
200 mg/kg, orally, for 7 days before LPS/DCL and twice (2 h and 8 h) after LPS/DCLTLR4/NF-κB/MAPK (p38/JNK) inhibition↓ TNF-α
↓ IL-6
↓ NO
[78]
Traumatic brain injury
C57BL/6 mice
50–200 mg/kg,
i.p.,
single dose
TLR4/MyD88/NF-κB inhibition↓ IL-1β
↓ IL-6
↓ TNF-α
↓ MCP-1
↓ RANTES
[70]
CCl4-induced hepatic fibrosis
Sprague-Dawley rats
200 mg/kg, orally, 6 weeksTLR4/HMGB1 inhibition↓ col3a1
↓ alpha-SMA
↓ TNF-a
↓ IL-6
↓ MCP-1
[79]
Triazole curcuminCurcuma longa LinnLPS-induced ALI
Kunming mice
2.5–20 mg/kg,
orally,
single dose, before LPS treatment
TLR4/MyD88/NF-κB/AP-1 inhibition↓ TNF-α
↓ IL-6
↓ fibrosis
[71]
QuercetinAllium cepa
Vitis vinifera
Solanum lycopersicum
Brassica oleracea
CCl4-induced hepatic inflammation
ICR mice
40 -80 mg/kg,
orally,
1 week
TLR4/NF-κB (p65)/MAPK (p38/JNK/ERK) inhibition↓ iNOS
↓ IL-1β
↓ COX-2
↓ NO
[76]
Concanavalin A– induced hepatitis
BALB/c mice
50 mg/kg, i.p., single dose
before concanavalin A
TLR2/HMGB1/NF-κB inhibition↓ TNF-α
↓ Interferon-γ
↓ IL-4
[80]
GlycyrrhizinGlycyrrhiza glabraLPS-induced ALI
BALB/c mice
50 mg/kg,
i.v.,
immediately and 12 h following LPS injection
TLR4/NF-κB inhibition↓ TNF-α
↓ IL-1α
↓ IL-6
↓ neutrophil and macrophage infiltration
↓ MPO
↓ COX-2
↓ iNOS
[81]
ResveratrolVitis viniferaLPS-induced ALI
BALB/c mice
5–45 mg/kg,
orally,
3 days before LPS injection
TLR4/MyD88/NF-κB inhibition↓ IL-6
↓ COX-2
[82]
SalidrosideRhodiola roseaLPS-induced ALI
Sprague–Dawley rats
20–40 mg/kg,
orally,
3 days
TLR4/NF-κB inhibition↓ TNF-α
↓ IL-6
↓ IL-1β
[83]
HesperetinCitrus spp.LPS-induced ALI
C57BL/6 mice
10–30 mg/kg,
orally,
single dose
TLR4/MyD88/TRAF6/TAK1/NF-κB (p65) inhibition; IκB-α stabilization↓ TNF-α
↓ IL-6
↓ NO
[84]
HesperidinCitrus spp.CLP-induced sepsis-associated lung injury
albino mice
10–20 mg/kg,
orally,
single dose
Hsp70/TLR4/MyD88 inhibition↓ TNF-α
↓ IL-6
↓ IL-1
[85]
AllicinAllium sativumLPS-induced ALI
Sprague-Dawley rats
25–100 μg/mL, i.p.,
every 12 h, for 24 h
TLR4/MyD88/NF-κB inhibition↓ TNF-α
↓ IL-6
↓ IL-1β
[86]
Alcohol-induced hepatic steatosis
C57BL/6 mice
5–20 mg/kg,
orally,
4 weeks
TLR4/CD14 inhibition↓ TNF-α
↓ IL-1β
↓ IL-6
[87]
MagnoflorineMagnolia spp.
Aristolochia spp.
LPS-induced ALI
BALB/c mice
5–20 mg/kg,
i.p.,
three times at 0, 8,
16 h after LPS injection
TLR4/NF-κB/MAPK (p38/ERK/JNK) inhibition; IκBα stabilization↓ TNF-α
↓ IL-1β
↓ IL-6
[88]
MagnololMagnolia officinalisLPS-induced ALI
BALB/c mice
5–20 mg/kg,
i.p.,
single dose
TLR4/NF-κB inhibition; IκBα stabilization↓ TNF-α
↓ IL-1β
↓ IL-6
[89]
ProcyanidinVitis viniferaTiO2 nanoparticle–induced hepatotoxicity
Albino rats
75 mg/kg,
orally,
30 days
TLR4/NIK/NF-κB inhibition↓ TNF-α[90]
Chlorogenic acidCamellia sinensis
Meum athamanticum
LPS-induced AKI
C57BL/6 mice
5–20 mg/kg,
i.p.,
single dose, 1h before LPS injection
TLR4/MyD88 inhibition; NF-κB (p65) inhibition; IκB-α stabilization↓ TNF-α
↓ IL-1β
↓ IL-6
[91]
PaeonolPaeonia moutan SimsLPS-induced AKI
BALB/c mice
12.5–50 mg/kg,
orally,
7 days
TLR4/IKKβ/NF-κB (p65) inhibition; IκBα stabilization↓ TNF-α
↓ IL-1β
↓ IL-6
[92]
AnthocyaninsMyrica rubraCerebral ischemia–reperfusion injury
ICR mice
100–300 mg/kg,
orally,
1 week
TLR4/NLRP3 inhibition↓ TNF-α
↓ IL-18
↓ caspase-1
↓ NO
[93]
BerberineCoptis chinensisCLP-induced sepsis
Wistar rats
25–50 mg/kg,
orally,
5 days
TLR4/NF-κB/NLRP3 inhibition↓ TNF-α
↓ IL-1β
[94]
LPS-induced sepsis-associated myocardial injury
Sprague Dawley rats
50 mg/kg,
orally,
3 days before LPS injection
TLR4/NF-κB (p65) inhibition↓ TNF-α
↓ IL-1β
[77]
Acute alcohol–induced gastrointestinal injury
ICR mice
75–300 mg/kg, orally,
1h before alcohol administration
TLR4 inhibition↓ TNF-α
↓ IL-1β
[95]
GenipinGardenia jasminoidesCLP–induced sepsis
ICR mice
1–5 mg/kg, i.v., immediately (0 h) or 0 and 24 h after CLP TLR2/HMGB1/MyD88/TRIF/NF-κB (p65)/MAPK (p38/JNK/ERK)/IRF3; IκBα stabilization↓ TNF-α
↓ IL-1β
↓ IL-6
[96]
Legend: ↓, decreased; ↑, increased; ALI, acute lung injury; alpha-SMA, alpha-smooth muscle actin; AKI, acute kidney injury; AP-1, activator protein-1; CCl4, carbon tetrachloride; CCL2, C-C motif chemokine ligand 2; CLP, cecal ligation and puncture; col3a1, type III collagen a 1; COX-2, cyclooxygenase-2; CXCL2, C-X-C motif chemokine ligand 2; D-GalN, D-galactosamine; DCL, diclofenac; ELAM, endothelial-leukocyte adhesion molecule; ERK1/2, extracellular signal-regulated kinase 1/2; F4/80+, macrophage marker F4/80-positive cells; HFD, high-fat diet; HMGB1, high mobility group box 1; Hsp70, heat shock protein 70; ICAM, intercellular adhesion molecule; ICR, Institute of Cancer Research (mouse strain); IFN-γ, interferon gamma; IL-1α, interleukin-1 alpha; IL-1β, interleukin-1 beta; IL-4, interleukin-4; IL-6, interleukin-6; IL-10, interleukin-10; IL-18, interleukin-18; iNOS, inducible nitric oxide synthase; IκB-α, inhibitor of nuclear factor kappa B alpha; IRAK1, interleukin-1 receptor-associated kinase 1; IRF3, interferon regulatory factor; JNK, c-Jun N-terminal kinase; MAP3K8, mitogen-activated protein kinase kinase kinase 8; MAPK, mitogen-activated protein kinase; MCD, methionine- and choline-deficient; MCP-1, monocyte chemoattractant protein-1; MPO, myeloperoxidase; MyD88, myeloid differentiation primary response 88; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; NF-κB, nuclear factor kappa B; NIK, NF-κB-inducing kinase; NLRP3, NOD-, LRR- and pyrin domain-containing protein 3; NO, nitric oxide; p38 MAPK, p38 mitogen-activated protein kinase; LPS, lipopolysaccharide; MCP-1, monocyte chemotactic protein 1; P-HFD, processed high-fat diet; RANTES, regulated on activation, normal T cell expressed and secreted; TAK1, transforming growth factor beta-activated kinase 1; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor alpha; TRAF6, TNF receptor-associated factor 6; TRIF, TIR-domain-containing adapter-inducing interferon-β; VCAM, vascular cell adhesion molecule.
Table 5. Natural compounds targeting TLR2 signaling pathways in experimental models of inflammation and organ injury.
Table 5. Natural compounds targeting TLR2 signaling pathways in experimental models of inflammation and organ injury.
Natural CompoundSourcesExperimental ModelMethod of AdministrationTLR2-Related MechanismMain OutcomesReference
CurcuminCurcuma longa LinnConcanavalin A–induced autoimmune hepatitis
BALB/c mice
200 mg/kg,
orally,
40 min before Concanavalin A injection
TLR2 inhibition↓ TNF-α
↓ IFN-γ
↑ IL-10
↓ Kupffer cell infiltration (F4/80+)
[72]
CCl4-induced hepatic fibrosis
Sprague-Dawley rats
200 mg/kg, orally, 6 weeksTLR2/HMGB1 inhibition↓ col3a1
↓ alpha-SMA
↓ TNF-a
↓ IL-6
↓ MCP-1
[79]
GlycyrrhizinGlycyrrhiza glabraLung ischemia–reperfusion injury
BALB/c mice
200 mg/kg,
i.p.,
single dose
TLR2/MyD88/NF-κB inhibition↓ IL-1β
↓ IL-6
[99]
QuercetinAllium cepa
Vitis vinifera
Solanum lycopersicum
Brassica oleracea
CCl4-induced hepatic inflammation
ICR mice
40–80 mg/kg,
orally,
1 week
TLR2/NF-κB (p65)/MAPK (p38/JNK/ERK) inhibition↓ iNOS
↓ IL-1β
↓ COX-2
↓ NO
[76]
Concanavalin A-induced hepatitis
BALB/c mice
50 mg/kg, i.p., single dose before concanavalin ATLR2/HMGB1/NF-κB (p65) inhibition; IκB stabilization↓ TNF-α
↓ Interferon-γ
↓ IL-4
[80]
Quercetin/Baicalein Quercetin:
Allium cepa
Vitis vinifera
Solanum lycopersicum
Brassica oleracea
Baicalein:
Lepisorus ussuriensis
Scutellaria prostrata
MCT-induced SOS
Sprague Dawley rats
40 mg/kg, intragastrical administration, twice at 6 h and 30 h after MCT administrationTLR2/MyD88/NF-κB (p-p65/nuclear-p65/p-IκB)/Egr1/MAPK (p-ASK1/p-MEK1/2/p-cRaf/p-MKK3/6/p-MKK4/p38/JNK/ERK 1/2)/PI3K/AKT/mTOR inhibition; Nrf2 activation↓ TNF-α
↓ IL-1β
↓ MDA
↓ MPO
↓ MMP-9
↓ Serpine1
↓ TF
↑ GCLC
↑ GCLM
[100]
PaeoniflorinPaeonia lactiflora PallType 2 diabetes-induced nephropathy
db/db mice
15–60 mg,
i.p.,
2 weeks
TLR2/MyD88/IRAK1/NF-κB/iNOS inhibition↓ TNF-α
↓ IL-1β
↓ MCP-1
↓ CD68+
[74]
BerberineCoptis chinensisAcute alcohol–induced gastrointestinal injury
ICR mice
75–300 mg/kg, orally,
1h before alcohol administration
TLR2 inhibition↓ TNF-α
↓ IL-1β
[95]
ResveratrolVitis viniferaCCl4–induced liver fibrosis
BALB/c mice
400 mg/kg.d, i.p., 5 weeks4 weeks: TLR2/MyD88/ERK/NF-κB (p50)/NLRP3
activation
5 weeks:
TLR2/MyD88/ERK/NF-κB (p50)/NLRP3
inhibition
4 weeks:
↑ IL-10
↑ Caspase-1
↑ IL-1β
↑ IL-18
5 weeks:
↓ IL-10
↓ Caspase-1
↓ IL-1β
↓ IL-18
[98]
GenipinGardenia jasminoidesCLP–induced sepsis
ICR mice
1–5 mg/kg, i.v., immediately (0 h) or 0 and 24 h after CLP TLR2/HMGB1/MyD88/TRIF/NF-κB (p65)/MAPK (p38/JNK/ERK)/IRF3; IκBα stabilization↓ TNF-α
↓ IL-1β
↓ IL-6
[96]
DioscinDioscorea
nipponica Makino
Zymosan-induced SIRS
C57BL/6J mice and Sprague Dawley rats
20–80 mg/kg for mice and 15–60 mg/kg for rats, orally, 7 daysTLR2/HMGB1/MyD88/NF-κb inhibition; IκBα stabilization↓ TNF-α
↓ IL-1β
↓ IL-6
↓ MPO
↓ MDA
↑ SOD
↓CD68+ macrophages
[97]
Legend: ↓, decreased; ↑, increased; AKT, protein kinase B; alpha-SMA, alpha-smooth muscle actin; CCl4, carbon tetrachloride; CLP, cecal ligation and puncture; col3a1, type III collagen a 1; COX-2, cyclooxygenase-2; cRaf, serine/threonine kinase in the Raf pathway; Egr1, early growth response1; ERK1/2, extracellular signal-regulated kinase 1/2; F4/80+, macrophage marker F4/80-positive cells; GCLC, glutamate-cysteine ligase catalytic subunit; GCLM, glutamate-cysteine ligase modifier subunit; HMGB1, high mobility group box 1; IFN-γ, interferon gamma; IL-1β, interleukin-1 beta; IL-4, interleukin-4; IL-6, interleukin-6; IL-10, interleukin-10; IL-18, interleukin-18; iNOS, inducible nitric oxide synthase; IκB, inhibitor of nuclear factor kappa B; IRAK1, interleukin-1 receptor-associated kinase 1; IRF3, interferon regulatory factor; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; MCP-1, monocyte chemoattractant protein-1; MCT, monocrotaline; MDA, malondialdehyde; MEK1/2, MAPK/ERK kinase 1/2; MKK3/6, MAPK kinase 3/6; MKK4, MAPK kinase 4; MMP9, matrix metalloproteinase 9; MPO, myeloperoxidase; MyD88, myeloid differentiation primary response 88; NF-κB, nuclear factor kappa B; NLRP3, Nucleotide-binding Oligomerization Domain-like Receptor Protein 3; NO, nitric oxide; Nrf2, nuclear factor erythroid 2–related factor 2; p38, p38 mitogen-activated protein kinase; LPS, lipopolysaccharide; PI3K, phosphoinositide 3-kinase; SIRS, Systemic inflammatory response syndrome; SOS, sinusoidal obstruction syndrome; TF, tissue factor; TLR2, toll-like receptor 2; TNF-α, tumor necrosis factor alpha; TRIF, TIR-domain-containing adapter-inducing interferon-β.
Table 6. Natural compounds targeting TLR9 signaling pathways in experimental models of DNA-sensing–driven inflammation and tissue injury.
Table 6. Natural compounds targeting TLR9 signaling pathways in experimental models of DNA-sensing–driven inflammation and tissue injury.
Natural CompoundSourcesExperimental ModelMethod of AdministrationTLR9-Related MechanismMain OutcomesReference
GlycyrrhizinGlycyrrhiza glabraCLP-induced sepsis-associated ARDS
C57BL/6 mice
15 mg/kg, i.p., single dose after CLPTLR9/HMGB1/MyD88 inhibition↓ Cit-H3
↓ NETs in lung tissue
↓ IL-6
[103]
CurcuminCurcuma longa LinnConcanavalin A–induced autoimmune hepatitis
BALB/c mice
200 mg/kg,
orally, single dose
40 min before Concanavalin A injection
TLR9 inhibition↓ TNF-α
↓ IFN-γ
↑ IL-10
↓ Kupffer cell infiltration (F4/80+)
[72]
CLP–induced AKI
Sprague-Dawley rats
40 mg/kg, orally, single dose after CLPTLR9/MYD88/IRF5/IRF7/NF-κB inhibition↓ TNF-α
↓ IL-10
↓ NGAL
↓ KIM-1
↓ CysC
[102]
Tanshinone IIASalvia miltiorrhiza BungeCollagen–induced arthritis
DBA/1 mice
5 mg/kg, orally, once a day from days 21 to 48 daysTLR9/RAGE/MMP9 inhibition↓ TNF-α
↓ IL-1β
↓ IL-6
[104]
Oleanolic acidOlea europaea
Salvia miltiorrhiza
Sambucus chinensis
STZ–induced diabetes
Sprague Dawley rats
5 mg/kg, orally, 21 daysTLR-9/NF-κB inhibition↓ IL-18
↓ MDA
[105]
OxymatrineDaphniphyllum spp.TNBS-induced colitis
Sprague-Dawley rats
10–60 mg/kg, i.p., 7 daysTLR9/Myd88/NF-κB (p65) inhibition↓ TNF-α
↓ IL-1β
↓ IL-6
↓ IL-10
↑ ZO-1
↑ occludin
↑ claudin-2
[101]
Quercetin/Baicalein Quercetin:
Allium cepa
Vitis vinifera
Solanum lycopersicum
Brassica oleracea
Baicalein:
Lepisorus ussuriensis
Scutellaria prostrata
MCT-induced SOS
Sprague Dawley rats
40 mg/kg, intragastrical administration, twice at 6 h and 30 h after MCT administrationTLR9/MyD88/NF-κB (p-p65/nuclear-p65/p-IκB)/Egr1/MAPK (p-ASK1/p-MEK1/2/p-cRaf/p-MKK3/6/p-MKK4/p38/JNK/ERK 1/2)/PI3K/AKT/mTOR inhibition; Nrf2 activation↓ TNF-α
↓ IL-1β
↓ MDA
↓ MPO
↓ MMP-9
↓ Serpine1
↓ TF
↑ GCLC
↑ GCLM
[100]
GastrodinGastrodia elataIschemic stroke injury (middle cerebral artery occlusion/reperfusion)
Sprague Dawley rats
25–100 mg/kg, i.p., 3 days before surgery and an additional 7 days post-surgery.mDNA/TLR9/JAK2/STAT3 inhibition↓ TNF-α
↓ IL-1β
↓ IL-6
↑ CAT
↑ GSH
↓ MDA
↑ SOD
↑ ATP
↑ T-ATPase
[106]
Betulinic acidPaeonia emodi
Bowdichia virgilioides
APAP-induced hepatotoxicity
Sprague Dawley rats
25 mg/kg, orally, 15 daysTLR-9/NF-κB inhibition↓ IL-18
↓ MDA
[107]
Legend: ↓, decreased; ↑, increased; AKI, acute kidney injury; AKT, protein kinase B; ARDS, acute respiratory distress syndrome; ASK1, apoptosis signal-regulating kinase 1; ATP, adenosine triphosphate; CAT, catalase; CysC, cystatin-C; Cit-H3, citrullinated histone 3; CLP, Cecal ligation and puncture; cRaf, serine/threonine kinase in the Raf pathway; Egr1, early growth response1; ERK1/2, extracellular signal-regulated kinase 1/2; GCLC, glutamate-cysteine ligase catalytic subunit; GCLM, glutamate-cysteine ligase modifier subunit; GSH, glutathione; HMGB1, high mobility group box 1; IκB, inhibitor of nuclear factor kappa B; IFN-γ, interferon gamma; IL-6, interleukin-6; IL-10, interleukin-10; IL-18, interleukin-18; IRF5, interferon regulatory factor 5; IRF7, interferon regulatory factor 7; JNK, c-Jun N-terminal kinase; KIM-1, kidney injury molecule-1; MAPK, mitogen-activated protein kinase; MCT, monocrotaline; MDA, malondialdehyde; mDNA, mitochondrial DNA; MEK1/2, MAPK/ERK kinase 1/2; MKK3/6, MAPK kinase 3/6; MKK4, MAPK kinase 4; MMP9, matrix metalloproteinase 9; MPO, myeloperoxidase; MyD88, myeloid differentiation primary response 88; mTOR, mechanistic target of rapamycin; NETs, neutrophil extracellular traps; NF-κB, nuclear factor kappa B; NGAL, neutrophil gelatinase-associated lipocalin; Nrf2, nuclear factor erythroid 2–related factor 2; PI3K, phosphoinositide 3-kinase; RAGE, receptor for advanced glycation end product; SOD, superoxide dismutase; SOS, sinusoidal obstruction syndrome; STZ, streptozotocin; TF, tissue factor; TLR9, toll-like receptor 9; TNBS, 2,4,6-trinitrobenzene sulfonic acid; TNF-α, tumor necrosis factor alpha; ZO-1, zonula occluden.
Table 7. Overview of natural TLR modulators: chemical classification, physicochemical characteristics, and safety information [108].
Table 7. Overview of natural TLR modulators: chemical classification, physicochemical characteristics, and safety information [108].
Natural CompoundChemical ClassPhysicochemical PropertiesSafety Data
BaicalinGlycosyloxyflavoneMW = 446.4 g/mol;
XLogP3 = 1.1;
TPSA = 183 Å2;
HBD = 6; HBA = 11
-
PaeoniflorinTerpene glycosideMW = 480.5 g/mol;
XLogP3 = −1;
TPSA = 164 Å2;
HBD = 5; HBA = 11
Mouse (i.p.) LD50 = 3530 mg/kg;
Mouse (i.v.) LD50 = 9530 mg/kg
Adverse effects: sleep, somnolence.
Rotundic acidTriterpenoidMW = 488.7 g/mol; XLogP3 = 5.3;
TPSA = 98 Å2;
HBD = 4; HBA = 5
-
Ginsenoside Rb1GinsenosideMW = 1109.3 g/mol;
XLogP3 = 0.3;
TPSA = 377 Å2;
HBD = 15; HBA = 23
Mouse (i.p.) LD50 = 1110 mg/kg; Mouse (i.v.) LD50 = 243 mg/kg
CurcuminBeta-diketoneMW = 368.4 g/mol;
XLogP3 = 3.2;
TPSA = 93.1 Å2;
HBD = 2; HBA = 6
Mouse (i.p.) LD50 = 1500 mg/kg; Mouse (oral) LD50 > 2 g/kg;
Rat (oral) LD50 > 5 g/kg;
Rabbit (dermal) LD50 > 5 g/kg.
QuercetinPentahydroxyflavoneMW = 302.23 g/mol;
XLogP3 = 1.5;
TPSA = 127 Å2;
HBD = 5; HBA = 7
Mouse (oral) LD50 = 159 mg/kg
Mouse (i.p.) LD50 = 3 g/kg
Mouse (s.c.) LD50 = 97 mg/kg
Rat (oral) LD50 = 161 mg/kg.
Adverse effects: somnolence, muscle weakness, respiratory depression;
GlycyrrhizinTriterpenoid saponinMW = 822.9 g/mol;
XLogP3 = 3.7;
TPSA = 267 Å2;
HBD = 8; HBA = 16
Human (oral) TDLo = 5571 µg/kg/3D—changes in urine
composition;
Human (oral) TDLo = 280 mg/kg/4 weeks—somnolence;
Human (oral) TDLo = 662 mg/kg/1 year—convulsions, muscle weakness;
Rat (oral) LDLo = 3 g/kg;
Rat (i.p.) LDLo = 2 g/kg;
Mouse (oral) LD50 = 4320 mg/kg;
Mouse (i.p.) LDLo = 1 g/kg; Mouse (i.v.) LD50 = 589 mg/kg—respiratory stimulation, other respiratory changes.
ResveratrolStilbeneMW = 228.24 g/mol;
XLogP3 = 3.1;
TPSA = 60.7 Å2;
HBD = 3; HBA = 3
Rat (oral, 90 days, feed) BMDL05 = 344 mg/kg bw/day –body weight decrease;
Human (oral, 1.5–3.0 g/day): mild, reversible ALT/AST increases
SalidrosideGlycosideMW = 300.30 g/mol;
XLogP3 = −0.6;
TPSA = 120 Å2;
HBD = 5; HBA = 7
Mouse (s.c.) LD50 = 28,600 µL/kg
HesperetinTrihydroxyflavanoneMW = 302.28 g/mol;
XLogP3 = 2.4;
TPSA = 96.2 Å2;
HBD = 3; HBA = 6
-
HesperidinFlavanone disaccharideMW = 610.6 g/mol;
XLogP3 = –1.1;
TPSA = 234 Å2;
HBD = 8; HBA = 15
Mouse (i.p.) LD50 = 1 g/kg
AllicinSulfoxideMW = 162.3 g/mol; XLogP3 = 1.3;
TPSA = 61.6 Å2;
HBD = 0; HBA = 3
Mouse (s.c.) LD50 = 120 mg/kg; Mouse (i.v.) LD50 = 60 mg/kg
MagnoflorineAlkaloidMW = 342.4 g/mol;
XLogP3 = 2.7;
TPSA = 58.9 Å2;
HBD = 2; HBA = 4
Mouse (i.p.) LD50 = 19,600 µg/kg; Mouse (s.c.) LD50 = 138 mg/kg; Mouse (i.v.) LD50 = 20 mg/kg
MagnololBiphenylMW = 266.3 g/mol;
XLogP3 = 5;
TPSA = 40.5 Å2;
HBD = 2; HBA = 2
Mouse (oral) LD50 = 2200 mg/kg
ProcyanidinProanthocyanidin oligomerMW = 594.5 g/mol;
XLogP3 = 2;
TPSA = 230 Å2;
HBD = 10; HBA = 13
-
Chlorogenic acidCinnamate esterMW = 354.31 g/mol;
XLogP3 = −0.4;
TPSA = 165 Å2;
HBD = 6; HBA = 9
Rat (i.p.) LDLo = 4 g/kg
PaeonolPhenolMW = 166.17 g/mol;
XLogP3 = 2;
TPSA = 46.5 Å2;
HBD = 1; HBA = 3
Mouse (oral) LD50 = 490 mg/kg;
Mouse (i.p.) LD50 = 781 mg/kg—altered sleep time;
Mouse (i.v.) LD50 = 196 mg/kg—altered sleep time
AnthocyaninsFlavonoidMW = 207.25 g/mol;
TPSA = 1 Å2;
HBD = 0; HBA = 0
-
BerberineAlkaloidMW = 336.4 g/mol;
XLogP3 = 3.6;
TPSA = 40.8 Å2;
HBD = 0; HBA = 4
Rat (i.p.) LD > 500 mg/kg;
Mouse (oral) LD50 = 329 mg/kg;
Mouse (s.c.) LD50 = 18 mg/kg;
Rabbit (s.c.) LDLo = 100 mg/kg.
GenipinIridoid monoterpenoidMW = 226.23 g/mol;
XLogP3 = −0.7;
TPSA = 76 Å2;
HBD = 2; HBA = 5
Mouse (oral) LD50 = 237 mg/kg;
Mouse (i.p.) LD50 = 190 mg/kg;
Mouse (i.v.) LD50 = 153 mg/kg.
BaicaleinTrihydroxyflavoneMW = 270.24 g/mol; XLogP3 = 1.7;
TPSA = 87 Å2;
HBD = 3; HBA = 5
-
DioscinSpirostanyl glycosideMW = 869.0 g/mol; XLogP3 = 1.3;
TPSA = 236 Å2;
HBD = 8; HBA = 16
Mouse (s.c.) LD50 > 300 mg/kg
Tanshinone IIADiterpenoidMW = 294.3 g/mol; XLogP3 = 4.3;
TPSA = 47.3 Å2;
HBD = 0; HBA = 3
-
Oleanolic acidTriterpenoidMW = 456.7 g/mol; XLogP3 = 7.5;
TPSA = 57.5 Å2;
HBD = 2; HBA = 3
Rat (oral) LD50 > 2 g/kg;
Rat (i.p.) LD50 > 2 g/kg;
Mouse (oral) LD50 > 2 g/kg;
Mouse (i.p.) LD50 = 1500 mg/kg
OxymatrineAlkaloidMW = 264.36 g/mol; XLogP3 = 1;
TPSA = 38.4 Å2;
HBD = 0; HBA = 2
Mouse (i.p.) LD50 = 521 mg/kg;
Mouse (i.m.) LD50 = 257 mg/kg;
Mouse (i.v.) LD50 = 150 mg/kg
GastrodinPhenolic glycosideMW = 286.28 g/mol; XLogP3 = −0.8;
TPSA = 120 Å2;
HBD = 5; HBA = 7
-
Betulinic acidTriterpenoidMW = 456.7 g/mol; XLogP3 = 8.2;
TPSA = 57.5 Å2;
HBD = 2; HBA = 3
-
Legend: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMDL05, benchmark dose lower confidence limit for 5% response; bw, body weight; HBA, hydrogen bond acceptors; HBD, hydrogen bond donors; i.m., intramuscular; i.p., intraperitoneal; i.v., intravenous; LDLo, lowest published lethal dose; LD50, median lethal dose; mg/kg, milligrams per kilogram; MW, molecular weight; s.c., subcutaneous; TDLo, lowest published toxic dose; TPSA, topological polar surface area; µg/kg, micrograms per kilogram; µL/kg, microlitres per kilogram; XLogP3, computed octanol–water partition coefficient. Chemical classification, physicochemical characteristics, and safety data were obtained from the PubChem database (National Center for Biotechnology Information) [108].
Table 8. Representative examples of natural compounds showing neutral or context-dependent effects on TLR signaling.
Table 8. Representative examples of natural compounds showing neutral or context-dependent effects on TLR signaling.
CompoundModel/CellTLR TargetAgonist/ContextEffect vs. Control
Chlorogenic acidPrimary rat hepatic stellate cellsTLR4/MyD88LPSNo change in TLR4 or MyD88 expression vs. control; downstream NF-κB/ROS inhibited [113]
GlycyrrhizinRAW264.7 macrophages and airway epithelium modelsTLR4/CD14LPS or HMGB1-rich supernatantsNo effect on TLR4/CD14 expression; reduces TLR4 translocation to lipid rafts and downstream signaling [114]
ResveratrolRAW264.7 macrophagesTLR2/3/4/9 pathwaysPoly I:C (TLR3), LPS (TLR4), Pam3CSK4 (TLR2), CpG (TLR9)Inhibits TRIF-dependent (TLR3/4) NF-κB, no inhibition on MyD88-dependent TLR2/9 [115]
BerberineLPS-stimulated macrophagesTLR4/MyD88LPSTLR4 protein unchanged; interferes with TLR4–MyD88 signaling [116]
CurcuminHepatocytes/marrow (ischemia–reperfusion in vivo and in vitro)TLR4/NF-κBH/R injury or LPSNo significant change vs. control in unstimulated groups; inhibits TLR4/NF-κB only under injurious stimulation [117]
Legend: CD14, Cluster of differentiation 14 (co-receptor for TLR4); CpG, Cytosine–phosphate–guanine oligodeoxynucleotides; HMGB1, High-mobility group box 1; H/R, Hypoxia/reoxygenation; LPS, Lipopolysaccharide; MyD88, Myeloid differentiation primary response 88 (TLR adaptor protein); NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; Pam3CSK4, Synthetic triacylated lipopeptide; Poly I:C, Polyinosinic–polycytidylic acid; ROS, Reactive oxygen species; TLR, Toll-like receptor; TRIF, TIR-domain-containing adapter-inducing interferon-β.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Andrei, C.; Pușcașu, C.; Nitulescu, G.M.; Zanfirescu, A. Modulation of Toll-like Receptors with Natural Compounds: A Therapeutic Avenue Against Inflammaging? Int. J. Mol. Sci. 2025, 26, 11305. https://doi.org/10.3390/ijms262311305

AMA Style

Andrei C, Pușcașu C, Nitulescu GM, Zanfirescu A. Modulation of Toll-like Receptors with Natural Compounds: A Therapeutic Avenue Against Inflammaging? International Journal of Molecular Sciences. 2025; 26(23):11305. https://doi.org/10.3390/ijms262311305

Chicago/Turabian Style

Andrei, Corina, Ciprian Pușcașu, George Mihai Nitulescu, and Anca Zanfirescu. 2025. "Modulation of Toll-like Receptors with Natural Compounds: A Therapeutic Avenue Against Inflammaging?" International Journal of Molecular Sciences 26, no. 23: 11305. https://doi.org/10.3390/ijms262311305

APA Style

Andrei, C., Pușcașu, C., Nitulescu, G. M., & Zanfirescu, A. (2025). Modulation of Toll-like Receptors with Natural Compounds: A Therapeutic Avenue Against Inflammaging? International Journal of Molecular Sciences, 26(23), 11305. https://doi.org/10.3390/ijms262311305

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop