Toll-like receptor 4

Last updated
TLR4
TLR4.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases TLR4 , ARMD10, CD284, TLR-4, TOLL, toll like receptor 4
External IDs OMIM: 603030 MGI: 96824 HomoloGene: 41317 GeneCards: TLR4
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_138557
NM_003266
NM_138554
NM_138556

NM_021297

RefSeq (protein)

NP_003257
NP_612564
NP_612567

NP_067272

Location (UCSC) Chr 9: 117.7 – 117.72 Mb Chr 4: 66.75 – 66.85 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Toll-like receptor 4 (TLR4), also designated as CD284 (cluster of differentiation 284), is a key activator of the innate immune response and plays a central role in the fight against bacterial infections. TLR4 is a transmembrane protein of approximately 95 kDa that is encoded by the TLR4 gene.

TLR4 belongs to the toll-like receptor family which is representative of the pattern recognition receptors (PRR), so named for their ability to recognize evolutionarily conserved components of microorganisms (bacteria, viruses, fungi and parasites) called pathogen-associated molecular patterns (PAMPs). The recognition of a PAMP by a PRR triggers rapid activation of the innate immunity essential to fight infectious diseases. [5]

TLR4 is expressed in immune cells mainly of myeloid origin, including monocytes, macrophages and dendritic cells (DC). [5] It is also expressed at a lower level on some non-immune cells, including epithelium, endothelium, placental cells and beta cells in Langerhans islets. Most myeloid cells express also high amounts of plasma membrane-anchored CD14, which facilitates the activation of TLR4 by LPS and controls the subsequent internalization of the LPS-activated TLR4 important for receptor signaling and degradation. [6] [7]

The main ligands for TLR4 are lipopolysaccharides (LPS), the major components of the outer membrane of Gram-negative bacteria and some Gram-positive bacteria. TLR4 can also be activated by endogenous compounds called damage-associated molecular patterns (DAMPs), including high mobility group box protein 1 (HMGB1), S100 proteins, or histones. These compounds are released during tissue injury and by dying or necrotic cells. [8] [9] [10] [11] [12]

Function

The first function described for TLR4 was the recognition of exogenous molecules from pathogens (PAMPs), in particular LPS molecules from gram-negative bacteria. [13] As pattern recognition receptor, TLR4 plays a fundamental role in pathogen recognition and activation of innate immunity which is the first line of defense against invading micro-organisms. During infection, TLR4 responds to the LPS present in tissues and the bloodstream and triggers pro-inflammatory reactions facilitating eradication of the invading bacteria. [13]

TLR4 is also involved in the recognition of endogenous DAMP molecules leading to different signaling outcomes than PAMPs, both quantitatively and qualitatively. [14] [12] DAMPs can activate TLR4 in non-infectious conditions to induce tissue repair and the activation of mainly proinflammatory responses. [8] [9] [10] [11] [12] Generally, inflammation has a protective role. It is a complex and coordinated process followed by the induction of resolution pathways that restore tissue integrity and function. However, in some cases, an excessive and/or poorly regulated inflammatory response to DAMPs can be detrimental to the organism, accelerating the development or progression of pathologies such as a number of cancers and neurodegenerative diseases (as discussed below).

TLR4 binds LPS with the help of LPS-binding protein (LBP) and CD14, and an indispensable contribution of the MD-2 protein stably associated with the extracellular fragment of the receptor. [15] TLR4 signaling responds to signals by forming a complex using an extracellular leucine-rich repeat domain (LRR) and an intracellular toll/interleukin-1 receptor (TIR) domain. LPS stimulation induces a series of interactions with several accessory proteins which form the TLR4 complex on the cell surface. LPS recognition is initiated by an LPS binding to an LBP protein. This LPS-LBP complex transfers the LPS to CD14 which is a glycosylphosphatidylinositol-anchored membrane protein that binds the LPS-LBP complex and facilitates the transfer of LPS to MD-2 protein, which is associated with the extracellular domain of TLR4. LPS binding promotes the dimerization of TLR4/MD-2 complex. The conformational changes of the TLR4 induce the recruitment of intracellular adaptor proteins containing the TIR domain which is necessary to activate the downstream signaling pathway.

The binding of an LPS molecule to the TLR4/MD-2 complex involves acyl chains and phosphate groups of lipid A, the conserved part of LPS and the main inducer of pro-inflammatory responses to LPS. [16] [17]

TLR4 activation and response to LPS is greatly influenced by the polysaccharide domain and the molecular structure of Lipid A moiety of the LPS molecules. Hexa-acylated and diphosphorylated LPS, like Escherichia coli LPS (O111:B4), is one of the most potent agonists of TLR4 whereas under-acylated LPS and dephosphorylated LPS species have a weaker pro-inflammatory activity especially in human cells. [18] Structural determinants of this phenomenon are found in the TLR4/MD-2 complex and also in CD14 protein. [16] [19] The polysaccharide portion covalently bound to lipid A plays also and indispensable role in TLR4 activation through CD14/TLR4/MD-2. [20] However, in addition to the lipid A domain, the polysaccharide moiety plays an important role in the binding and activation of the LPS molecules as the lipid A moiety alone was demonstrated to be significantly less active than the full LPS molecule. [21]

Signaling

Unlike all the other TLRs, TLR4 stimulation triggers two signaling pathways called the MyD88-dependent and the TRIF-dependent one after the adaptor proteins involved in their induction. [22] The MyD88-dependent signaling is triggered by TLR4 localized to the plasma membrane, while the TRIF-dependent one by the TLR4 internalized in endosomes.

These signaling pathways lead to the production of two sets of cytokines. The MyD88-dependent pathway induces the production of pro-inflammatory cytokines while TRIF-dependent pathway induces the production of type I interferons and chemokines. [22] [23] The molecular structure of TLR4 ligands (in particular LPS), as well as their complexation with proteins or lipids, greatly influence the action of these TLR4-related signaling pathways, leading to different cytokine balances. [24] [25] [26] [27]

MyD88 and TRIF dependent signaling pathway of TLR4. TLR4 signaling pathways V2.pdf
MyD88 and TRIF dependent signaling pathway of TLR4.

MyD88 – dependent pathway

The MyD88-dependent pathway is regulated by two adaptor-associated proteins: Myeloid Differentiation Primary Response Gene 88 (MyD88) and TIR Domain-Containing Adaptor Protein (TIRAP). It also involves the activation of IL-1 Receptor-Associated Kinases (IRAKs) and the adaptor molecules TNF Receptor-Associated Factor 6 (TRAF6). TRAF6 induces the activation of TAK1 (Transforming growth factor-β-Activated Kinase 1) that leads to the activation of MAPK cascades (Mitogen-Activated Protein Kinase) and the IκB Kinases (IKK), called IKKα and IKKβ. [28] IKKs' signaling pathway leads to the induction of the transcription factor NF-κB, while activation of MAPK cascades lead to the activation of another transcription factor AP-1. [28] [29] These two transcription factorsnduces the expression of genes encoding pro-inflammatory mediators, such as tumor necrosis factor α (TNF-α), interleukin (IL)-6, and type III interferons (IFNλ1/2). [30] [31] [32]

TRIF – dependent pathway

The TRIF-dependent pathway involves the internalization of TLR4 in endosomes and the recruitment of the adaptor proteins TIR-domain-containing adaptor inducing interferon-β (TRIF) and TRIF-related Adaptor Molecule (TRAM). TRAM-TRIF signals activate the ubiquitin ligase TRAF3 followed by the activation of non-canonical IKK kinases: TANK binding kinase 1 (TBK1) and IKKε. TBK1 phosphorylates the pLxIS consensus motif of TRIF that is necessary to recruit interferon regulatory factor (IRF) 3. IRF3 is also phosphorylated by TBK1 and then dissociates from TRIF, dimerizes and translocates to the nucleus. [33] Finally, IRF3 induces the expression of genes encoding type I IFN such as interferon beta (IFN-β), the chemokine CCL5/ RANTES and interferon-regulated genes as that encoding the chemokine CXCL10/IP-10. [30] [31] [32] [34] TRIF-dependent signaling pathway of TLR4 is known to play a central role in the stimulation of innate immune cells such as macrophages, the maturation of DCs and the induction and recruitment of Th1 adaptive immune responses. [35]

Immune cell activation

TLR4 activation by LPS enables a rapid stimulation of a whide range of innate immune cells such as macrophages and DCs. This leads to the secretion of pro-inflammatory and type I interferons cytokines, chemokines. Production levels of these cytokines/chemokines vary according to the degree of activation of the MyD88 and TRIF signaling pathways by TLR4 agonist molecules. TLR4 activation also induces the stimulation of antigen presentation and upregulation of costimulatory molecules (such as CD40, CD80 and CD86) on innate immune cells which are required for antigen presentation for T lymphocytes. [36] [37] This explains why TLR4 activation by LPS is also known to stimulate the generation of effective adaptive immune responses and to induce their recruitment, polarization and maintenance via the panel of cytokines and chemokines produced. [37] [22]

The TRIF and MyD88 signaling pathways have a different but complementary impact on immune cell activation. Macrophages stimulation has been shown to be strictly dependent on TRIF pathway activation whereas DC activation and maturation depend on both the MyD88 and TRIF pathways. [38] [39] [40] [41] The increased expression of costimulatory and MHC molecules is a hallmark of DC maturation required for antigen presentation by these cells. [42]


However, significant differences were found in the signaling pathways leading to this phenomenon. In macrophages, the upregulation of costimulatory molecules depends strictly on the TRIF-dependent pathway, whereas in DC both the MyD88- and TRIF-dependent ones are involved. [43] [44] [22] [45] The increased cell surface presence of the costimulatory molecules and also of MHC II is a hallmark of DC maturation required for antigen presentation by these cells. [46]

The activation of MyD88 and TRIF signaling pathways were also found to induce Th1 polarization of the T cells responses through DC maturation and the panel of cytokines produced. [47] [48] [49] Low activation of MYD88 pathway is however important for effective cytotoxic T-cell differentiation by facilitating fusion of MHC I-bearing recycling endosomes with phagosomes allowing cross-presentation of antigens. [47] In contrast, robust activation of MYD88 pathway induces excessive production of pro-inflammatory cytokines leading to life-threatening pathological consequences such as cytokine storms.

The impact of TLR4 activation on the innate and adaptive immune system explains why TLR4 agonists, such as LPS derivatives, have been developed as vaccine adjuvants. Among them is GSK's Monophosphorylated Lipid A (MPL), a detoxified Lipid A derived from Salmonella LPS, which is the first and only natural immunostimulant to have been approved as adjuvant in five human vaccines. [50] [51] [52]

Evolutionary history and polymorphism

TLR4 originated when TLR2 and TLR4 diverged about 500 million years ago near the beginning of vertebrate evolution. [53] Sequence alignments of human and great ape TLR4 exons have demonstrated that not much evolution has occurred in human TLR4 since our divergence from our last common ancestor with chimpanzees; human and chimp TLR4 exons only differ by three substitutions while humans and baboons are 93.5% similar in the extracellular domain. [54] Notably, humans possess a greater number of early stop codons in TLR4 than great apes; in a study of 158 humans worldwide, 0.6% had a nonsense mutation. [55] [56] This suggests that there are weaker evolutionary pressures on the human TLR4 than on our primate relatives. The distribution of human TLR4 polymorphisms matches the out-of-Africa migration, and it is likely that the polymorphisms were generated in Africa before migration to other continents. [56] [57]

Various single nucleotide polymorphisms (SNPs) of TLR4 have been identified in humans . For some of them, an association with increased susceptibility to Gram-negative bacterial infections or faster progression and a more severe course of sepsis in critically ill patients was reported.However, they are very rare, and their frequency varies according to ethnic origin. The 2 predominant SNPs are Asp299Gly and Thr399Ile, with a frequency of <10% in the Caucasian population and even lower in the Asian population. [58] These two SNPs are missense mutations, thus associated with a loss of function, which may explain their negative impact on infection control. Studies have indeed shown that TLR4 D299G SNP limits the response to LPS by compromising MyD88 and TRIF recruitment to TLR4, and thus cytokine secretion, but without affecting TLR4 expression [59] [60] Structural analyses of human TLR4 with SNP D299G suggest that this amino acid change affects van der Waals interaction and hydrogen bonding in leucine-rich repeats, modulating its surface properties which may affect LPS ligand binding to TLR4. [61]

Clinical significance

TLR4 has been reported to play both friend and foe in a variety of human diseases, such as bacterial infections and cancers. This dual role of TLR4 depends on the intensity, duration and site (surface or endosome) of its activation, its polymorphism and the balance of activation of signaling pathways (MyD88 vs. TRIF).

Infectious diseases

TLR4 play a central role in the control of bacterial infections through the recognition of LPS molecules from gram-negative, and some gram-positive, bacteria. [62] During infections, TLR4s on innate immunity cells are activated by LPS molecules present in tissues and the bloodstream. This activates innate immunity, the first line of defense against invading microorganisms, and triggers pro-inflammatory responses that facilitate the eradication of invading bacteria. [13] Generally, inflammation has a protective role. It is a complex and coordinated process followed by the induction of resolution pathways that restore tissue integrity and function. However, in some cases, exaggerated and uncontrolled inflammation triggered by TLR4 during infection can lead to sepsis and septic shock. [33] Infections with Gram-negative bacteria such as Escherichia coli and Pseudomonas aeruginosa are the prevailing causes of severe sepsis in humans.Some studies have linked TLR4 polymorphisms (Asp299Gly and Thr399Ile SNPs) to an increased susceptibility to sepsis due to gram-negative infection but other studies failed to confirm this. [63]

Cancer

The role of the TLR4 in the control of cancer progression and in cancer therapy is well documented.

Stimulation of TLR4 by natural derivatives and LPS is well known to induce potent antitumor activity. This anti-tumor activity is linked to the ability of LPS to stimulate innate immunity via TLR4, resulting in the production of pro-inflammatory cytokines and type 1 interferons, and the indirect generation of adaptive anti-tumor responses. [64] [65]

The first clues about the efficacy of TLR4 agonists like LPS in cancer immunotherapy was found in the 19th centuries, when bacterial infections were found to induce tumor regressions. [66] Later, Dr William Coley showed the therapeutic efficacy of a mixed bacterial vaccine, so-called “Coley’s toxin”, to human cancer. [67] Since then, a number of developments have been made in the treatment or prevention of cancer using bacterial mixtures strongly activating TLR4 due to LPS content. The antituberculosis vaccine Bacillus Calmette–Guérin (BCG) was approved by the Federal Drug Administration (FDA) in 1990 for the local treatment of superficial bladder cancer. BCG promotes dendritic cell maturation, and this effect is TLR4 (as well as TLR2) dependent. [68] There are also reports on the treatment of oral squamous cell carcinoma, gastric , Head-and-neck and cervical cancers with lyophilized streptococcal preparation OK-432 (Picibanil). [69] The mechanism of action of OK-432 involves TLR4 activation, since OKA-432 does not inhibit tumor growth on TLR4 knockouts as it does on wild-type mice. [70]

Purified LPS also showed potent anti-tumor efficacy as systemic therapeutic agents in several tumor models. [71] [72] In the 90’s, clinical trials evaluating the intravenous administration of LPS to patients with cancer provided positive results including several cases of disease stabilization and partial responses. However, limiting toxicities at doses in the ng/kg range has been reported which are too low to obtain significant antitumor effects. [73]

Subsequently, detoxified TLR4 agonists (LPS derivatives) have been produced and evaluated in the clinic. This includes the MPL, a chemically modified LPS which was the first TLR4 agonist to be approved and commercialized by GSK in 5 human vaccines (HPV, Zoster, Hepatitis B, Malaria, RSV). MPL was investigated as an adjuvant for curative anti-tumor vaccines, with the approval of Melacine in Canada for the treatment of patients with malignant melanoma. [74] Synthetic LPS derivatives based on dephosphorylated lipid A moiety structures were also developed and confirmed potent adjuvant and antitumor activities as therapeutic agents. In particular, the intratumoral administration of Glucopyranosyl Lipid Adjuvant (GLA-SE/G100), a synthetic detoxified analog of lipid A formulated in a stable emulsion, showed anti-tumor immune responses and tumor regression in patients with Merkel cell carcinoma, [75] and potent adjuvant activity in phase 2 trials in combination with pembrolizumab in patients with follicular lymphoma. [76] [77]

Besides the recognized anti-tumor efficacy of TLR4 activation by LPS, some studies suggest that TLR4 may also contribute to the development of some cancers, (prostate, liver, breast and lung cancers) and may contribute to resistance to paclitaxel chemotherapy in breast cancer. [78] Some clinical studies also suggested a potential correlation between TLR4 expression on tumor cells and tumor progression. However, no such effect was reported in the numerous clinical studies conducted with natural LPS or LPS derivatives. On the contrary, in phase 2 studies with GLA, a positive association between baseline TLR4 expression in tumors and the increase of overall response rates has been reported. [77]

The potential impact of TLR4 on the progression of some cancers was associated with the excessive production of pro-inflammatory cytokines via activation of the TLR4-MyD88/NF-kB signaling pathway. [79] [80] [81] Several studies showed that this is mediated by the misuse of DAMP signaling by tumor cells. [12] [82] [14]

Many DAMPs are released by dying or necrotic tumor cells and present during cancer progression. DAMPs released from tumor cells can directly activate tumor-expressed TLR4 that induce chemoresistance, migration, invasion, and metastasis. Furthermore, DAMP-induced chronic inflammation in the tumor microenvironment causes an increase in immunosuppressive populations, such as M2 macrophages, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs). [12] DAMPs, such as HMGB1, S100 proteins, and heat shock proteins (HSPs), were found to strongly activate inflammatory pathways and release IL-1, IL-6, LT-β, IFN-γ, TNF, and transforming growth factor (TGF)-β promoting inflammation, immunosuppression, angiogenesis, and tumor cell proliferation. [11]

Several studies have evaluated the potential association of this TLR4 polymorphism with cancer risk, but the data are highly conflicting. However, some meta-analyses suggest an association of SNP D299G with gastric, viral-induced and female-specific cancers (cervix, ovary). [83]

Neurogenerative diseases

Growing evidence suggests an implication of TLR4 in the development and progression of neurogenerative disorders such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. In the brain, TLR4 is expressed by neurons as well as the non-neuronal glial cells, which include microglia, astrocytes, and oligodendrocytes. TLR4 is expressed primarily by microglia, and to a lesser extent by astrocytes, oligodendrocytes, and neurons. [5] Microglia are representatives of the mononuclear phagocyte system in the brain, and TLR4 activation regulates some of their functions, such as phagocytic activity. [84] [13]

Activation of microglial TLR4 has been suggested to protect against or slow the development of neurodegenerative diseases, notably by enhancing the clearance of neurotoxic proteins such as Aβ and its aggregates, thanks to increased phagocytic and autophagic activity. [85]

However, chronic TLR4 activation is believed to be associated with glia-mediated neuronal death due to excessive secretion of pro-inflammatory cytotoxins leading to neuroinflammation, a key factor in the development of many neurodegenerative diseases. [86] [87] In the brain, TLR4 can be activated by various endogenous DAMPs in addition to pathology-associated proteins such as aggregates of amyloid-βpeptides (Aβ) or α-synuclein. [88] All these structures bind TLR4 and activate downstream signaling pathways in glia, inducing secretion of reactive oxygen species (ROS) and proinflammatory cytokines such as IL-1β and TNF-α, which can lead to damage and death of neurons. [86] [89] [90] Neuronal death is accompanied by the release of DAMPs into the extracellular space, which can then further activate TLR4, aggravating neuroinflammation. [91] In patients with Alzheimer's disease (AD), the levels of circulating DAMPs like HMGB1 and soluble RAGE, are significantly elevated, which was correlated with the levels of amyloid beta. [92] In AD patients, the serum levels of S100B are also intimately related to the severity of the disease. [93] The role of the HMGB1-TLR4 axis is very important in the pathogenesis of Parkinson's disease (PD). The serum HMGB1 and TLR4 protein levels were significantly elevated in PD patients and correlated with the PD stages. [94]

Targeting TLR4 with agonists or antagonists, or modulating its downstream signaling pathways, may have a therapeutic potential in treating neurodegenerative diseases. [95] TLR4-specific antagonists could suppress neuroinflammation by reducing overproduction of inflammatory mediators and cytotoxins by glia. However, TLR4 antagonists could have adverse CNS effects by inhibiting phagocytosis by glia, reducing protein clearance, and interfering with myelination. [96] Some studies showed that selective TLR4 agonists could be beneficial by upregulating the phagocytic activity of microglia, leading to enhanced clearance of damaged tissue and abnormal protein aggregates associated with several different CNS diseases. Repeated injections of MPL, at doses that are nonpyrogenic, were found to significantly improved AD-related pathology mice. [97] MPL led to a significant reduction in Aβ load in the brain, as well as enhanced cognitive function. MPL induced a potent phagocytic response by microglia while triggering a moderate inflammatory reaction. However, adverse effects can be caused by TLR 4 agonists inducing secretion of inflammatory mediators. Studies therefore suggested that TLR4 agonists that selectively activate the TRIF signaling pathway could be highly beneficial in the treatment of neurodegenerative disorders by increasing glial cell phagocytic activity without significantly increasing glial cytokines and cytotoxins. [96]

Drugs targeting TLR4

TLR4 has been shown to be important for the long-term side-effects of opioid analgesic drugs. Various μ-opioid receptor ligands have been tested and found to also possess action as agonists or antagonists of TLR4, with opioid agonists such as (+)-morphine being TLR4 agonists, while opioid antagonists such as naloxone were found to be TLR4 antagonists. Activation of TLR4 leads to downstream release of inflammatory modulators including TNF-α and Interleukin-1, and constant low-level release of these modulators is thought to reduce the efficacy of opioid drug treatment with time, and be involved in both the development of tolerance to opioid analgesic drugs, [98] [99] and in the emergence of side-effects such as hyperalgesia and allodynia that can become a problem following extended use of opioid drugs. [100] [101] Drugs that block the action of TNF-α or IL-1β have been shown to increase the analgesic effects of opioids and reduce the development of tolerance and other side-effects, [102] [103] and this has also been demonstrated with drugs that block TLR4 itself.

The response of TLR4 to opioid drugs has been found to be enantiomer-independent, so the "unnatural" enantiomers of opioid drugs such as morphine and naloxone, which lack affinity for opioid receptors, still produce the same activity at TLR4 as their "normal" enantiomers. [104] [105] This means that the unnatural enantiomers of opioid antagonists, such as (+)-naloxone, can be used to block the TLR4 activity of opioid analgesic drugs, while leaving the μ-opioid receptor mediated analgesic activity unaffected. [106] [105] [107] This may also be the mechanism behind the beneficial effect of ultra-low dose naltrexone on opioid analgesia. [108]

Morphine causes inflammation by binding to the protein lymphocyte antigen 96, which, in turn, causes the protein to bind to Toll-like receptor 4 (TLR4). [109] The morphine-induced TLR4 activation attenuates pain suppression by opioids and enhances the development of opioid tolerance and addiction, drug abuse, and other negative side effects such as respiratory depression and hyperalgesia. Drug candidates that target TLR4 may improve opioid-based pain management therapies. [110]

Agonists

Apart from LPS and its derivatives, up to 30 natural TLR4 agonists with diverse chemical structures have been postulated. However, besides DAMPs, the others have not demonstrated to be direct activators of TLR4 and could therefore act as chaperones for TLR4 or as promoters of LPS internalization. [8] [111] [112]

Antagonists

As of 2020, there were no specific TLR4 antagonists approved as drugs. [113]

Related Research Articles

<span class="mw-page-title-main">Natural killer cell</span> Type of cytotoxic lymphocyte

Natural killer cells, also known as NK cells or large granular lymphocytes (LGL), are a type of cytotoxic lymphocyte critical to the innate immune system. They belong to the rapidly expanding family of known innate lymphoid cells (ILC) and represent 5–20% of all circulating lymphocytes in humans. The role of NK cells is analogous to that of cytotoxic T cells in the vertebrate adaptive immune response. NK cells provide rapid responses to virus-infected cells, stressed cells, tumor cells, and other intracellular pathogens based on signals from several activating and inhibitory receptors. Most immune cells detect the antigen presented on major histocompatibility complex I (MHC-I) on infected cell surfaces, but NK cells can recognize and kill stressed cells in the absence of antibodies and MHC, allowing for a much faster immune reaction. They were named "natural killers" because of the notion that they do not require activation to kill cells that are missing "self" markers of MHC class I. This role is especially important because harmful cells that are missing MHC I markers cannot be detected and destroyed by other immune cells, such as T lymphocyte cells.

<span class="mw-page-title-main">Toll-like receptor</span> Class of immune system proteins

Toll-like receptors (TLRs) are a class of proteins that play a key role in the innate immune system. They are single-spanning receptors usually expressed on sentinel cells such as macrophages and dendritic cells, that recognize structurally conserved molecules derived from microbes. Once these microbes have reached physical barriers such as the skin or intestinal tract mucosa, they are recognized by TLRs, which activate immune cell responses. The TLRs include TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, and TLR13. Humans lack genes for TLR11, TLR12 and TLR13 and mice lack a functional gene for TLR10. The receptors TLR1, TLR2, TLR4, TLR5, TLR6, and TLR10 are located on the cell membrane, whereas TLR3, TLR7, TLR8, and TLR9 are located in intracellular vesicles.

Pathogen-associated molecular patterns (PAMPs) are small molecular motifs conserved within a class of microbes, but not present in the host. They are recognized by toll-like receptors (TLRs) and other pattern recognition receptors (PRRs) in both plants and animals. This allows the innate immune system to recognize pathogens and thus, protect the host from infection.

Pattern recognition receptors (PRRs) play a crucial role in the proper function of the innate immune system. PRRs are germline-encoded host sensors, which detect molecules typical for the pathogens. They are proteins expressed mainly by cells of the innate immune system, such as dendritic cells, macrophages, monocytes, neutrophils, as well as by epithelial cells, to identify two classes of molecules: pathogen-associated molecular patterns (PAMPs), which are associated with microbial pathogens, and damage-associated molecular patterns (DAMPs), which are associated with components of host's cells that are released during cell damage or death. They are also called primitive pattern recognition receptors because they evolved before other parts of the immune system, particularly before adaptive immunity. PRRs also mediate the initiation of antigen-specific adaptive immune response and release of inflammatory cytokines.

<span class="mw-page-title-main">IRAK4</span> Protein-coding gene in humans

IRAK-4, in the IRAK family, is a protein kinase involved in signaling innate immune responses from Toll-like receptors. It also supports signaling from T-cell receptors. IRAK4 contains domain structures which are similar to those of IRAK1, IRAK2, IRAKM and Pelle. IRAK4 is unique compared to IRAK1, IRAK2 and IRAKM in that it functions upstream of the other IRAKs, but is more similar to Pelle in this trait. IRAK4 has important clinical applications.

<span class="mw-page-title-main">MYD88</span> Protein found in humans

Myeloid differentiation primary response 88 (MYD88) is a protein that, in humans, is encoded by the MYD88 gene. originally discovered in the laboratory of Dan A. Liebermann as a Myeloid differentiation primary response gene.

<span class="mw-page-title-main">TICAM1</span> Protein found in humans

TIR domain containing adaptor molecule 1 is an adapter in responding to activation of toll-like receptors (TLRs). It mediates the rather delayed cascade of two TLR-associated signaling cascades, where the other one is dependent upon a MyD88 adapter.

<span class="mw-page-title-main">Toll-like receptor 7</span> Protein found in humans

Toll-like receptor 7, also known as TLR7, is a protein that in humans is encoded by the TLR7 gene. Orthologs are found in mammals and birds. It is a member of the toll-like receptor (TLR) family and detects single stranded RNA.

<span class="mw-page-title-main">Toll-like receptor 5</span> Protein found in humans

Toll-like receptor 5, also known as TLR5, is a protein which in humans is encoded by the TLR5 gene. It is a member of the toll-like receptor (TLR) family. TLR5 is known to recognize bacterial flagellin from invading mobile bacteria. It has been shown to be involved in the onset of many diseases, which includes Inflammatory bowel disease. Recent studies have also shown that malfunctioning of TLR5 is likely related to rheumatoid arthritis, osteoclastogenesis, and bone loss. Abnormal TLR5 functioning is related to the onset of gastric, cervical, endometrial and ovarian cancers.

<span class="mw-page-title-main">Lymphocyte antigen 96</span> Protein-coding gene in the species Homo sapiens

Lymphocyte antigen 96, also known as "Myeloid Differentiation factor 2 (MD-2)," is a protein that in humans is encoded by the LY96 gene.

<span class="mw-page-title-main">Toll-like receptor 6</span> Protein found in humans

Toll-like receptor 6 is a protein that in humans is encoded by the TLR6 gene. TLR6 is a transmembrane protein, member of toll-like receptor family, which belongs to the pattern recognition receptor (PRR) family. TLR6 acts in a heterodimer form with toll-like receptor 2 (TLR2). Its ligands include multiple diacyl lipopeptides derived from gram-positive bacteria and mycoplasma and several fungal cell wall saccharides. After dimerizing with TLR2, the NF-κB intracellular signalling pathway is activated, leading to a pro-inflammatory cytokine production and activation of innate immune response. TLR6 has also been designated as CD286.

<span class="mw-page-title-main">Toll-like receptor 9</span> Protein found in humans

Toll-like receptor 9 is a protein that in humans is encoded by the TLR9 gene. TLR9 has also been designated as CD289. It is a member of the toll-like receptor (TLR) family. TLR9 is an important receptor expressed in immune system cells including dendritic cells, macrophages, natural killer cells, and other antigen presenting cells. TLR9 is expressed on endosomes internalized from the plasma membrane, binds DNA, and triggers signaling cascades that lead to a pro-inflammatory cytokine response. Cancer, infection, and tissue damage can all modulate TLR9 expression and activation. TLR9 is also an important factor in autoimmune diseases, and there is active research into synthetic TLR9 agonists and antagonists that help regulate autoimmune inflammation.

<span class="mw-page-title-main">IRAK1</span> Protein-coding gene in humans

Interleukin-1 receptor-associated kinase 1 (IRAK-1) is an enzyme in humans encoded by the IRAK1 gene. IRAK-1 plays an important role in the regulation of the expression of inflammatory genes by immune cells, such as monocytes and macrophages, which in turn help the immune system in eliminating bacteria, viruses, and other pathogens. IRAK-1 is part of the IRAK family consisting of IRAK-1, IRAK-2, IRAK-3, and IRAK-4, and is activated by inflammatory molecules released by signaling pathways during pathogenic attack. IRAK-1 is classified as a kinase enzyme, which regulates pathways in both innate and adaptive immune systems.

<span class="mw-page-title-main">Bruce Beutler</span> American immunologist and geneticist

Bruce Alan Beutler is an American immunologist and geneticist. Together with Jules A. Hoffmann, he received one-half of the 2011 Nobel Prize in Physiology or Medicine, for "discoveries concerning the activation of innate immunity." Beutler discovered the long-elusive receptor for lipopolysaccharide. He did so by identifying spontaneous mutations in the gene coding for mouse Toll-like receptor 4 (Tlr4) in two unrelated strains of LPS-refractory mice and proving they were responsible for that phenotype. Subsequently, and chiefly through the work of Shizuo Akira, other TLRs were shown to detect signature molecules of most infectious microbes, in each case triggering an innate immune response.

<span class="mw-page-title-main">SIGIRR</span> Protein-coding gene in the species Homo sapiens

Single Ig IL-1-related receptor (SIGIRR), also called Toll/Interleukin-1 receptor 8 (TIR8) or Interleukin-1 receptor 8 (IL-1R8), is transmembrane protein encoded by gene SIGIRR, which modulate inflammation, immune response, and tumorigenesis of colonic epithelial cells.

<span class="mw-page-title-main">TICAM2</span> Protein-coding gene in the species Homo sapiens

TIR domain-containing adapter molecule 2 is a protein that in humans is encoded by the TICAM2 gene.

<span class="mw-page-title-main">Toll-interleukin receptor</span> Intracellular signaling domain

The toll-interleukin-1 receptor (TIR) homology domain is an intracellular signaling domain found in MyD88, SARM1, interleukin-1 receptors, toll receptors and many plant R proteins. It contains three highly conserved regions, and mediates protein-protein interactions between the toll-like receptors (TLRs) and signal-transduction components. TIR-like motifs are also found in plant proteins where they are involved in resistance to disease and in bacteria where they are associated with virulence. When activated, TIR domains recruit cytoplasmic adaptor proteins MyD88 (UniProt Q99836) and TOLLIP (toll-interacting protein, UniProt Q9H0E2). In turn, these associate with various kinases to set off signaling cascades. Some TIR domains have also been found to have intrinsic NAD+ cleavage activity, such as in SARM1. In the case of SARM1, the TIR NADase activity leads to the production of Nam, ADPR and cADPR and the activation of downstream pathways involved in Wallerian degeneration and neuron death.

Murine caspase-11, and its human homologs caspase-4 and caspase-5, are mammalian intracellular receptor proteases activated by TLR4 and TLR3 signaling during the innate immune response. Caspase-11, also termed the non-canonical inflammasome, is activated by TLR3/TLR4-TRIF signaling and directly binds cytosolic lipopolysaccharide (LPS), a major structural element of Gram-negative bacterial cell walls. Activation of caspase-11 by LPS is known to cause the activation of other caspase proteins, leading to septic shock, pyroptosis, and often organismal death.

The interleukin-1 receptor (IL-1R) associated kinase (IRAK) family plays a crucial role in the protective response to pathogens introduced into the human body by inducing acute inflammation followed by additional adaptive immune responses. IRAKs are essential components of the Interleukin-1 receptor signaling pathway and some Toll-like receptor signaling pathways. Toll-like receptors (TLRs) detect microorganisms by recognizing specific pathogen-associated molecular patterns (PAMPs) and IL-1R family members respond the interleukin-1 (IL-1) family cytokines. These receptors initiate an intracellular signaling cascade through adaptor proteins, primarily, MyD88. This is followed by the activation of IRAKs. TLRs and IL-1R members have a highly conserved amino acid sequence in their cytoplasmic domain called the Toll/Interleukin-1 (TIR) domain. The elicitation of different TLRs/IL-1Rs results in similar signaling cascades due to their homologous TIR motif leading to the activation of mitogen-activated protein kinases (MAPKs) and the IκB kinase (IKK) complex, which initiates a nuclear factor-κB (NF-κB) and AP-1-dependent transcriptional response of pro-inflammatory genes. Understanding the key players and their roles in the TLR/IL-1R pathway is important because the presence of mutations causing the abnormal regulation of Toll/IL-1R signaling leading to a variety of acute inflammatory and autoimmune diseases.

Jonathan C. Kagan is an American immunologist and the Marian R. Neutra, Ph.D. Professor of Pediatrics at Harvard Medical School. He is also the director of Basic Research and Shwachman Chair in Gastroenterology at Boston Children's Hospital. Kagan is a world leader in defining the molecular basis of innate immunity and inflammation.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000136869 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000039005 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. 1 2 3 Vaure C, Liu Y (2014). "A comparative review of toll-like receptor 4 expression and functionality in different animal species". Frontiers in Immunology. 5: 316. doi: 10.3389/fimmu.2014.00316 . PMC   4090903 . PMID   25071777.
  6. Mahnke K, Becher E, Ricciardi-Castagnoli P, Luger TA, Schwarz T, Grabbe S (1997). "CD14 is Expressed by Subsets of Murine Dendritic Cells and Upregulated by Lipopolysaccharide". In Ricciardi-Castagnoli P (ed.). Dendritic Cells in Fundamental and Clinical Immunology. Advances in Experimental Medicine and Biology. Vol. 417. Boston, MA: Springer US. pp. 145–159. doi:10.1007/978-1-4757-9966-8_25. ISBN   978-1-4757-9968-2. PMID   9286353.
  7. Sabroe I, Jones EC, Usher LR, Whyte MK, Dower SK (May 2002). "Toll-like receptor (TLR)2 and TLR4 in human peripheral blood granulocytes: a critical role for monocytes in leukocyte lipopolysaccharide responses". Journal of Immunology. 168 (9): 4701–4710. doi:10.4049/jimmunol.168.9.4701. PMID   11971020.
  8. 1 2 3 Yang H, Wang H, Ju Z, Ragab AA, Lundbäck P, Long W, et al. (January 2015). "MD-2 is required for disulfide HMGB1-dependent TLR4 signaling". The Journal of Experimental Medicine. 212 (1): 5–14. doi:10.1084/jem.20141318. PMC   4291531 . PMID   25559892.
  9. 1 2 Jiang D, Liang J, Fan J, Yu S, Chen S, Luo Y, et al. (November 2005). "Regulation of lung injury and repair by Toll-like receptors and hyaluronan". Nature Medicine. 11 (11): 1173–1179. doi:10.1038/nm1315. PMID   16244651. S2CID   11765495.
  10. 1 2 Fang H, Ang B, Xu X, Huang X, Wu Y, Sun Y, et al. (March 2014). "TLR4 is essential for dendritic cell activation and anti-tumor T-cell response enhancement by DAMPs released from chemically stressed cancer cells". Cellular & Molecular Immunology. 11 (2): 150–159. doi:10.1038/cmi.2013.59. PMC   4003380 . PMID   24362470.
  11. 1 2 3 Hernandez C, Huebener P, Schwabe RF (November 2016). "Damage-associated molecular patterns in cancer: a double-edged sword". Oncogene. 35 (46): 5931–5941. doi:10.1038/onc.2016.104. PMC   5119456 . PMID   27086930.
  12. 1 2 3 4 5 Jang GY, Lee JW, Kim YS, Lee SE, Han HD, Hong KJ, et al. (December 2020). "Interactions between tumor-derived proteins and Toll-like receptors". Experimental & Molecular Medicine. 52 (12): 1926–1935. doi:10.1038/s12276-020-00540-4. PMC   8080774 . PMID   33299138.
  13. 1 2 3 4 Molteni M, Gemma S, Rossetti C (2016). "The Role of Toll-Like Receptor 4 in Infectious and Noninfectious Inflammation". Mediators of Inflammation. 2016: 6978936. doi: 10.1155/2016/6978936 . PMC   4887650 . PMID   27293318.
  14. 1 2 Roh JS, Sohn DH (August 2018). "Damage-Associated Molecular Patterns in Inflammatory Diseases". Immune Network. 18 (4): e27. doi:10.4110/in.2018.18.e27. PMC   6117512 . PMID   30181915.
  15. Tsukamoto H, Takeuchi S, Kubota K, Kobayashi Y, Kozakai S, Ukai I, et al. (June 2018). "Lipopolysaccharide (LPS)-binding protein stimulates CD14-dependent Toll-like receptor 4 internalization and LPS-induced TBK1-IKKϵ-IRF3 axis activation". The Journal of Biological Chemistry. 293 (26): 10186–10201. doi: 10.1074/jbc.M117.796631 . PMC   6028956 . PMID   29760187.
  16. 1 2 Park BS, Song DH, Kim HM, Choi BS, Lee H, Lee JO (April 2009). "The structural basis of lipopolysaccharide recognition by the TLR4-MD-2 complex". Nature. 458 (7242): 1191–1195. Bibcode:2009Natur.458.1191P. doi:10.1038/nature07830. PMID   19252480. S2CID   4396446.
  17. Park BS, Lee JO (December 2013). "Recognition of lipopolysaccharide pattern by TLR4 complexes". Experimental & Molecular Medicine. 45 (12): e66. doi:10.1038/emm.2013.97. PMC   3880462 . PMID   24310172.
  18. Steimle A, Autenrieth IB, Frick JS (August 2016). "Structure and function: Lipid A modifications in commensals and pathogens". International Journal of Medical Microbiology. 306 (5): 290–301. doi: 10.1016/j.ijmm.2016.03.001 . PMID   27009633.
  19. Kelley SL, Lukk T, Nair SK, Tapping RI (February 2013). "The crystal structure of human soluble CD14 reveals a bent solenoid with a hydrophobic amino-terminal pocket". Journal of Immunology. 190 (3): 1304–1311. doi:10.4049/jimmunol.1202446. PMC   3552104 . PMID   23264655.
  20. Muroi M, Tanamoto K (November 2002). "The polysaccharide portion plays an indispensable role in Salmonella lipopolysaccharide-induced activation of NF-kappaB through human toll-like receptor 4". Infection and Immunity. 70 (11): 6043–6047. doi:10.1128/IAI.70.11.6043-6047.2002. PMC   130318 . PMID   12379680.
  21. Cavaillon JM, Fitting C, Caroff M, Haeffner-Cavaillon N (March 1989). "Dissociation of cell-associated interleukin-1 (IL-1) and IL-1 release induced by lipopolysaccharide and lipid A". Infection and Immunity. 57 (3): 791–797. doi:10.1128/iai.57.3.791-797.1989. PMC   313178 . PMID   2537258.
  22. 1 2 3 4 Shen H, Tesar BM, Walker WE, Goldstein DR (August 2008). "Dual signaling of MyD88 and TRIF is critical for maximal TLR4-induced dendritic cell maturation". Journal of Immunology. 181 (3): 1849–1858. doi:10.4049/jimmunol.181.3.1849. PMC   2507878 . PMID   18641322.
  23. Nakayama M, Niki Y, Kawasaki T, Takeda Y, Ikegami H, Toyama Y, et al. (October 2013). "IL-32-PAR2 axis is an innate immunity sensor providing alternative signaling for LPS-TRIF axis". Scientific Reports. 3 (1): 2960. Bibcode:2013NatSR...3E2960N. doi:10.1038/srep02960. PMC   3797434 . PMID   24129891.
  24. Pridmore AC, Jarvis GA, John CM, Jack DL, Dower SK, Read RC (July 2003). "Activation of toll-like receptor 2 (TLR2) and TLR4/MD2 by Neisseria is independent of capsule and lipooligosaccharide (LOS) sialylation but varies widely among LOS from different strains". Infection and Immunity. 71 (7): 3901–3908. doi:10.1128/IAI.71.7.3901-3908.2003. PMC   161978 . PMID   12819075.
  25. Stephenson HN, John CM, Naz N, Gundogdu O, Dorrell N, Wren BW, et al. (July 2013). "Campylobacter jejuni lipooligosaccharide sialylation, phosphorylation, and amide/ester linkage modifications fine-tune human Toll-like receptor 4 activation". The Journal of Biological Chemistry. 288 (27): 19661–19672. doi: 10.1074/jbc.M113.468298 . PMC   3707672 . PMID   23629657.
  26. Alexander-Floyd J, Bass AR, Harberts EM, Grubaugh D, Buxbaum JD, Brodsky IE, et al. (August 2022). Bäumler AJ (ed.). "Lipid A Variants Activate Human TLR4 and the Noncanonical Inflammasome Differently and Require the Core Oligosaccharide for Inflammasome Activation". Infection and Immunity. 90 (8): e0020822. doi:10.1128/iai.00208-22. PMC   9387229 . PMID   35862709.
  27. Bonhomme D, Santecchia I, Vernel-Pauillac F, Caroff M, Germon P, Murray G, et al. (August 2020). "Leptospiral LPS escapes mouse TLR4 internalization and TRIF‑associated antimicrobial responses through O antigen and associated lipoproteins". PLOS Pathogens. 16 (8): e1008639. doi: 10.1371/journal.ppat.1008639 . PMC   7447051 . PMID   32790743.
  28. 1 2 Pålsson-McDermott EM, O'Neill LA (October 2004). "Signal transduction by the lipopolysaccharide receptor, Toll-like receptor-4". Immunology. 113 (2): 153–162. doi:10.1111/j.1365-2567.2004.01976.x. PMC   1782563 . PMID   15379975.
  29. Lu YC, Yeh WC, Ohashi PS (May 2008). "LPS/TLR4 signal transduction pathway". Cytokine. 42 (2): 145–151. doi:10.1016/j.cyto.2008.01.006. PMID   18304834.
  30. 1 2 Meissner F, Scheltema RA, Mollenkopf HJ, Mann M (April 2013). "Direct proteomic quantification of the secretome of activated immune cells". Science. 340 (6131): 475–478. Bibcode:2013Sci...340..475M. doi:10.1126/science.1232578. PMID   23620052. S2CID   40513139.
  31. 1 2 Kawai T, Takeuchi O, Fujita T, Inoue J, Mühlradt PF, Sato S, et al. (November 2001). "Lipopolysaccharide stimulates the MyD88-independent pathway and results in activation of IFN-regulatory factor 3 and the expression of a subset of lipopolysaccharide-inducible genes". Journal of Immunology. 167 (10): 5887–5894. doi:10.4049/jimmunol.167.10.5887. PMID   11698465.
  32. 1 2 Chanteux H, Guisset AC, Pilette C, Sibille Y (October 2007). "LPS induces IL-10 production by human alveolar macrophages via MAPKinases- and Sp1-dependent mechanisms". Respiratory Research. 8 (1): 71. doi: 10.1186/1465-9921-8-71 . PMC   2080632 . PMID   17916230.
  33. 1 2 Ciesielska A, Matyjek M, Kwiatkowska K (February 2021). "TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling". Cellular and Molecular Life Sciences. 78 (4): 1233–1261. doi:10.1007/s00018-020-03656-y. PMC   7904555 . PMID   33057840.
  34. O'Neill LA, Golenbock D, Bowie AG (June 2013). "The history of Toll-like receptors - redefining innate immunity". Nature Reviews. Immunology. 13 (6): 453–460. doi:10.1038/nri3446. hdl: 2262/72552 . PMID   23681101. S2CID   205491986.
  35. Watanabe S, Kumazawa Y, Inoue J (2013). "Liposomal lipopolysaccharide initiates TRIF-dependent signaling pathway independent of CD14". PLOS ONE. 8 (4): e60078. Bibcode:2013PLoSO...860078W. doi: 10.1371/journal.pone.0060078 . PMC   3615118 . PMID   23565187.
  36. Lien E, Means TK, Heine H, Yoshimura A, Kusumoto S, Fukase K, et al. (February 2000). "Toll-like receptor 4 imparts ligand-specific recognition of bacterial lipopolysaccharide". The Journal of Clinical Investigation. 105 (4): 497–504. doi:10.1172/JCI8541. PMC   289161 . PMID   10683379.
  37. 1 2 Shetab Boushehri MA, Lamprecht A (November 2018). "TLR4-Based Immunotherapeutics in Cancer: A Review of the Achievements and Shortcomings". Molecular Pharmaceutics. 15 (11): 4777–4800. doi:10.1021/acs.molpharmaceut.8b00691. PMID   30226786. S2CID   52297047.
  38. Kaisho T, Takeuchi O, Kawai T, Hoshino K, Akira S (May 2001). "Endotoxin-induced maturation of MyD88-deficient dendritic cells". Journal of Immunology. 166 (9): 5688–5694. doi:10.4049/jimmunol.166.9.5688. PMID   11313410.
  39. Hoebe K, Janssen EM, Kim SO, Alexopoulou L, Flavell RA, Han J, et al. (December 2003). "Upregulation of costimulatory molecules induced by lipopolysaccharide and double-stranded RNA occurs by Trif-dependent and Trif-independent pathways". Nature Immunology. 4 (12): 1223–1229. doi:10.1038/ni1010. PMID   14625548. S2CID   8505015.
  40. Shen H, Tesar BM, Walker WE, Goldstein DR (August 2008). "Dual signaling of MyD88 and TRIF is critical for maximal TLR4-induced dendritic cell maturation". Journal of Immunology. 181 (3): 1849–1858. doi:10.4049/jimmunol.181.3.1849. PMC   2507878 . PMID   18641322.
  41. Trombetta ES, Ebersold M, Garrett W, Pypaert M, Mellman I (February 2003). "Activation of lysosomal function during dendritic cell maturation". Science. 299 (5611): 1400–1403. doi:10.1126/science.1080106. PMID   12610307. S2CID   46594244.
  42. Turley SJ, Inaba K, Garrett WS, Ebersold M, Unternaehrer J, Steinman RM, et al. (April 2000). "Transport of peptide-MHC class II complexes in developing dendritic cells". Science. 288 (5465): 522–527. doi:10.1126/science.288.5465.522. PMID   10775112.
  43. Kaisho T, Takeuchi O, Kawai T, Hoshino K, Akira S (May 2001). "Endotoxin-induced maturation of MyD88-deficient dendritic cells". Journal of Immunology. 166 (9): 5688–5694. doi:10.4049/jimmunol.166.9.5688. PMID   11313410.
  44. Hoebe K, Janssen EM, Kim SO, Alexopoulou L, Flavell RA, Han J, et al. (December 2003). "Upregulation of costimulatory molecules induced by lipopolysaccharide and double-stranded RNA occurs by Trif-dependent and Trif-independent pathways". Nature Immunology. 4 (12): 1223–1229. doi:10.1038/ni1010. PMID   14625548. S2CID   8505015.
  45. Trombetta ES, Ebersold M, Garrett W, Pypaert M, Mellman I (February 2003). "Activation of lysosomal function during dendritic cell maturation". Science. 299 (5611): 1400–1403. doi:10.1126/science.1080106. PMID   12610307. S2CID   46594244.
  46. Turley SJ, Inaba K, Garrett WS, Ebersold M, Unternaehrer J, Steinman RM, et al. (April 2000). "Transport of peptide-MHC class II complexes in developing dendritic cells". Science. 288 (5465): 522–527. doi:10.1126/science.288.5465.522. PMID   10775112.
  47. 1 2 Nair-Gupta P, Baccarini A, Tung N, Seyffer F, Florey O, Huang Y, et al. (July 2014). "TLR signals induce phagosomal MHC-I delivery from the endosomal recycling compartment to allow cross-presentation". Cell. 158 (3): 506–521. doi:10.1016/j.cell.2014.04.054. PMC   4212008 . PMID   25083866.
  48. Han JE, Wui SR, Kim KS, Cho YJ, Cho WJ, Lee NG (2014-01-22). Shin EC (ed.). "Characterization of the structure and immunostimulatory activity of a vaccine adjuvant, de-O-acylated lipooligosaccharide". PLOS ONE. 9 (1): e85838. Bibcode:2014PLoSO...985838H. doi: 10.1371/journal.pone.0085838 . PMC   3899070 . PMID   24465739.
  49. Sharif O, Bolshakov VN, Raines S, Newham P, Perkins ND (January 2007). "Transcriptional profiling of the LPS induced NF-kappaB response in macrophages". BMC Immunology. 8 (1): 1. doi: 10.1186/1471-2172-8-1 . PMC   1781469 . PMID   17222336.
  50. Paavonen J, Jenkins D, Bosch FX, Naud P, Salmerón J, Wheeler CM, et al. (June 2007). "Efficacy of a prophylactic adjuvanted bivalent L1 virus-like-particle vaccine against infection with human papillomavirus types 16 and 18 in young women: an interim analysis of a phase III double-blind, randomised controlled trial". Lancet. 369 (9580): 2161–2170. doi:10.1016/S0140-6736(07)60946-5. PMID   17602732. S2CID   26318328.
  51. Kundi M (April 2007). "New hepatitis B vaccine formulated with an improved adjuvant system". Expert Review of Vaccines. 6 (2): 133–140. doi:10.1586/14760584.6.2.133. PMID   17408363. S2CID   35472093.
  52. Garçon N, Di Pasquale A (January 2017). "From discovery to licensure, the Adjuvant System story". Human Vaccines & Immunotherapeutics. 13 (1): 19–33. doi:10.1080/21645515.2016.1225635. PMC   5287309 . PMID   27636098.
  53. Beutler B, Rehli M (2002). "Evolution of the TIR, Tolls and TLRS: Functional Inferences from Computational Biology". Toll-Like Receptor Family Members and Their Ligands. Current Topics in Microbiology and Immunology. Vol. 270. pp. 1–21. doi:10.1007/978-3-642-59430-4_1. ISBN   978-3-642-63975-3. PMID   12467241.
  54. Smirnova I, Poltorak A, Chan EK, McBride C, Beutler B (2000). "Phylogenetic variation and polymorphism at the toll-like receptor 4 locus (TLR4)". Genome Biology. 1 (1): RESEARCH002. doi: 10.1186/gb-2000-1-1-research002 . PMC   31919 . PMID   11104518.
  55. Quach H, Wilson D, Laval G, Patin E, Manry J, Guibert J, et al. (December 2013). "Different selective pressures shape the evolution of Toll-like receptors in human and African great ape populations". Human Molecular Genetics. 22 (23): 4829–4840. doi:10.1093/hmg/ddt335. PMC   3820138 . PMID   23851028.
  56. 1 2 Barreiro LB, Ben-Ali M, Quach H, Laval G, Patin E, Pickrell JK, et al. (July 2009). "Evolutionary dynamics of human Toll-like receptors and their different contributions to host defense". PLOS Genetics. 5 (7): e1000562. doi: 10.1371/journal.pgen.1000562 . PMC   2702086 . PMID   19609346.
  57. Plantinga TS, Ioana M, Alonso S, Izagirre N, Hervella M, Joosten LA, et al. (2012). "The evolutionary history of TLR4 polymorphisms in Europe". Journal of Innate Immunity. 4 (2): 168–175. doi:10.1159/000329492. PMC   6741577 . PMID   21968286.
  58. Noreen M, Shah MA, Mall SM, Choudhary S, Hussain T, Ahmed I, et al. (March 2012). "TLR4 polymorphisms and disease susceptibility". Inflammation Research. 61 (3): 177–188. doi:10.1007/s00011-011-0427-1. PMID   22277994. S2CID   9500302.
  59. Long H, O'Connor BP, Zemans RL, Zhou X, Yang IV, Schwartz DA (2014-04-02). "The Toll-like receptor 4 polymorphism Asp299Gly but not Thr399Ile influences TLR4 signaling and function". PLOS ONE. 9 (4): e93550. Bibcode:2014PLoSO...993550L. doi: 10.1371/journal.pone.0093550 . PMC   3973565 . PMID   24695807.
  60. Figueroa L, Xiong Y, Song C, Piao W, Vogel SN, Medvedev AE (May 2012). "The Asp299Gly polymorphism alters TLR4 signaling by interfering with recruitment of MyD88 and TRIF". Journal of Immunology. 188 (9): 4506–4515. doi:10.4049/jimmunol.1200202. PMC   3531971 . PMID   22474023.
  61. Ohto U, Yamakawa N, Akashi-Takamura S, Miyake K, Shimizu T (November 2012). "Structural analyses of human Toll-like receptor 4 polymorphisms D299G and T399I". The Journal of Biological Chemistry. 287 (48): 40611–40617. doi: 10.1074/jbc.M112.404608 . PMC   3504774 . PMID   23055527.
  62. Akira S, Takeda K (July 2004). "Toll-like receptor signalling". Nature Reviews. Immunology. 4 (7): 499–511. doi:10.1038/nri1391. PMID   15229469.
  63. Netea MG, Wijmenga C, O'Neill LA (May 2012). "Genetic variation in Toll-like receptors and disease susceptibility". Nature Immunology. 13 (6): 535–542. doi:10.1038/ni.2284. PMID   22610250. S2CID   24438756.
  64. Chettab K, Fitzsimmons C, Novikov A, Denis M, Phelip C, Mathé D, et al. (2023). "A systemically administered detoxified TLR4 agonist displays potent antitumor activity and an acceptable tolerance profile in preclinical models". Frontiers in Immunology. 14: 1066402. doi: 10.3389/fimmu.2023.1066402 . PMC   10200957 . PMID   37223101.
  65. Richert I, Berchard P, Abbes L, Novikov A, Chettab K, Vandermoeten A, et al. (September 2023). "A TLR4 Agonist Induces Osteosarcoma Regression by Inducing an Antitumor Immune Response and Reprogramming M2 Macrophages to M1 Macrophages". Cancers. 15 (18): 4635. doi: 10.3390/cancers15184635 . PMC   10526955 . PMID   37760603.
  66. Maruyama K, Selmani Z, Ishii H, Yamaguchi K (March 2011). "Innate immunity and cancer therapy". International Immunopharmacology. 11 (3): 350–357. doi:10.1016/j.intimp.2010.09.012. PMID   20955832.
  67. Starnes CO (May 1992). "Coley's toxins in perspective". Nature. 357 (6373): 11–12. Bibcode:1992Natur.357...11S. doi:10.1038/357011a0. PMID   1574121. S2CID   4265230.
  68. Tsuji S, Matsumoto M, Takeuchi O, Akira S, Azuma I, Hayashi A, et al. (December 2000). Kaufmann SH (ed.). "Maturation of human dendritic cells by cell wall skeleton of Mycobacterium bovis bacillus Calmette-Guérin: involvement of toll-like receptors". Infection and Immunity. 68 (12): 6883–6890. doi:10.1128/IAI.68.12.6883-6890.2000. PMC   97794 . PMID   11083809.
  69. Ryoma Y, Moriya Y, Okamoto M, Kanaya I, Saito M, Sato M (2004-09-01). "Biological effect of OK-432 (picibanil) and possible application to dendritic cell therapy". Anticancer Research. 24 (5C): 3295–3301. PMID   15515424.
  70. Okamoto M, Oshikawa T, Tano T, Ohe G, Furuichi S, Nishikawa H, et al. (February 2003). "Involvement of Toll-like receptor 4 signaling in interferon-gamma production and antitumor effect by streptococcal agent OK-432". Journal of the National Cancer Institute. 95 (4): 316–326. doi:10.1093/jnci/95.4.316. PMID   12591988.
  71. Shear MB, Perrault M (April 1944). "Chemical Treatment of Tumors. IX. Reactions of Mice with Primary Subcutaneous Tumors to Injection of a Hemorrhage-Producing Bacterial Polysaccharide1". JNCI: Journal of the National Cancer Institute. 4 (5): 461–476. doi:10.1093/jnci/4.5.461.
  72. Berendt MJ, North RJ, Kirstein DP (December 1978). "The immunological basis of endotoxin-induced tumor regression. Requirement for a pre-existing state of concomitant anti-tumor immunity". The Journal of Experimental Medicine. 148 (6): 1560–1569. doi:10.1084/jem.148.6.1560. PMC   2185097 . PMID   309922.
  73. Engelhardt R, Mackensen A, Galanos C (May 1991). "Phase I trial of intravenously administered endotoxin (Salmonella abortus equi) in cancer patients". Cancer Research. 51 (10): 2524–2530. PMID   2021932.
  74. "Melacine - an overview | ScienceDirect Topics". www.sciencedirect.com. Retrieved 2024-02-20.
  75. Bhatia S, Miller NJ, Lu H, Longino NV, Ibrani D, Shinohara MM, et al. (February 2019). "Intratumoral G100, a TLR4 Agonist, Induces Antitumor Immune Responses and Tumor Regression in Patients with Merkel Cell Carcinoma". Clinical Cancer Research. 25 (4): 1185–1195. doi:10.1158/1078-0432.CCR-18-0469. PMC   6368904 . PMID   30093453.
  76. Halwani AS, Panizo C, Isufi I, Herrera AF, Okada CY, Cull EH, et al. (April 2022). "Phase 1/2 study of intratumoral G100 (TLR4 agonist) with or without pembrolizumab in follicular lymphoma". Leukemia & Lymphoma. 63 (4): 821–833. doi:10.1080/10428194.2021.2010057. PMID   34865586. S2CID   244943266.
  77. 1 2 Flowers C, Panizo C, Isufi I, Herrera AF, Okada C, Cull EH, et al. (2017-12-08). "Intratumoral G100 Induces Systemic Immunity and Abscopal Tumor Regression in Patients with Follicular Lymphoma: Results of a Phase 1/ 2 Study Examining G100 Alone and in Combination with Pembrolizumab". Blood. 130: 2771. doi:10.1182/blood.V130.Suppl_1.2771.2771 (inactive 2024-02-20). ISSN   0006-4971.{{cite journal}}: CS1 maint: DOI inactive as of February 2024 (link)
  78. Rajput S, Volk-Draper LD, Ran S (August 2013). "TLR4 is a novel determinant of the response to paclitaxel in breast cancer". Molecular Cancer Therapeutics. 12 (8): 1676–1687. doi:10.1158/1535-7163.MCT-12-1019. PMC   3742631 . PMID   23720768.
  79. Zhang R, Zhao J, Xu J, Jiao DX, Wang J, Gong ZQ, et al. (October 2017). "Andrographolide suppresses proliferation of human colon cancer SW620 cells through the TLR4/NF-κB/MMP-9 signaling pathway". Oncology Letters. 14 (4): 4305–4310. doi:10.3892/ol.2017.6669. PMC   5604146 . PMID   28943944.
  80. Wang CH, Wang PJ, Hsieh YC, Lo S, Lee YC, Chen YC, et al. (February 2018). "Resistin facilitates breast cancer progression via TLR4-mediated induction of mesenchymal phenotypes and stemness properties". Oncogene. 37 (5): 589–600. doi:10.1038/onc.2017.357. PMID   28991224. S2CID   24926622.
  81. Kelly MG, Alvero AB, Chen R, Silasi DA, Abrahams VM, Chan S, et al. (April 2006). "TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer". Cancer Research. 66 (7): 3859–3868. doi:10.1158/0008-5472.CAN-05-3948. PMID   16585214.
  82. Khademalhosseini M, Arababadi MK (May 2019). "Toll-like receptor 4 and breast cancer: an updated systematic review". Breast Cancer. 26 (3): 265–271. doi:10.1007/s12282-018-00935-2. PMID   30543015. S2CID   56143069.
  83. Zhu L, Yuan H, Jiang T, Wang R, Ma H, Zhang S (2013-12-20). "Association of TLR2 and TLR4 polymorphisms with risk of cancer: a meta-analysis". PLOS ONE. 8 (12): e82858. Bibcode:2013PLoSO...882858Z. doi: 10.1371/journal.pone.0082858 . PMC   3869723 . PMID   24376595.
  84. Wardill HR, Van Sebille YZ, Mander KA, Gibson RJ, Logan RM, Bowen JM, et al. (February 2015). "Toll-like receptor 4 signaling: a common biological mechanism of regimen-related toxicities: an emerging hypothesis for neuropathy and gastrointestinal toxicity". Cancer Treatment Reviews. 41 (2): 122–128. doi:10.1016/j.ctrv.2014.11.005. PMID   25512119.
  85. Tahara K, Kim HD, Jin JJ, Maxwell JA, Li L, Fukuchi K (November 2006). "Role of toll-like receptor signalling in Abeta uptake and clearance". Brain. 129 (Pt 11): 3006–3019. doi:10.1093/brain/awl249. PMC   2445613 . PMID   16984903.
  86. 1 2 Buchanan MM, Hutchinson M, Watkins LR, Yin H (July 2010). "Toll-like receptor 4 in CNS pathologies". Journal of Neurochemistry. 114 (1): 13–27. doi:10.1111/j.1471-4159.2010.06736.x. PMC   2909662 . PMID   20402965.
  87. Qin Y, Liu Y, Hao W, Decker Y, Tomic I, Menger MD, et al. (October 2016). "Stimulation of TLR4 Attenuates Alzheimer's Disease-Related Symptoms and Pathology in Tau-Transgenic Mice". Journal of Immunology. 197 (8): 3281–3292. doi:10.4049/jimmunol.1600873. PMID   27605009.
  88. Gambuzza M, Licata N, Palella E, Celi D, Foti Cuzzola V, Italiano D, et al. (October 2011). "Targeting Toll-like receptors: emerging therapeutics for multiple sclerosis management". Journal of Neuroimmunology. 239 (1–2): 1–12. doi:10.1016/j.jneuroim.2011.08.010. PMID   21889214. S2CID   3277551.
  89. Rannikko EH, Weber SS, Kahle PJ (September 2015). "Exogenous α-synuclein induces toll-like receptor 4 dependent inflammatory responses in astrocytes". BMC Neuroscience. 16: 57. doi: 10.1186/s12868-015-0192-0 . PMC   4562100 . PMID   26346361.
  90. Walter S, Letiembre M, Liu Y, Heine H, Penke B, Hao W, et al. (2007). "Role of the toll-like receptor 4 in neuroinflammation in Alzheimer's disease". Cellular Physiology and Biochemistry. 20 (6): 947–956. doi:10.1159/000110455. PMID   17982277. S2CID   6752610.
  91. Land WG (February 2015). "The Role of Damage-Associated Molecular Patterns in Human Diseases: Part I - Promoting inflammation and immunity". Sultan Qaboos University Medical Journal. 15 (1): e9–e21. PMC   4318613 . PMID   25685392.
  92. Festoff BW, Sajja RK, van Dreden P, Cucullo L (August 2016). "HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease". Journal of Neuroinflammation. 13 (1): 194. doi: 10.1186/s12974-016-0670-z . PMC   4995775 . PMID   27553758.
  93. Chaves ML, Camozzato AL, Ferreira ED, Piazenski I, Kochhann R, Dall'Igna O, et al. (January 2010). "Serum levels of S100B and NSE proteins in Alzheimer's disease patients". Journal of Neuroinflammation. 7: 6. doi: 10.1186/1742-2094-7-6 . PMC   2832635 . PMID   20105309.
  94. Yang Y, Han C, Guo L, Guan Q (April 2018). "High expression of the HMGB1-TLR4 axis and its downstream signaling factors in patients with Parkinson's disease and the relationship of pathological staging". Brain and Behavior. 8 (4): e00948. doi:10.1002/brb3.948. PMC   5893335 . PMID   29670828.
  95. Wu L, Xian X, Xu G, Tan Z, Dong F, Zhang M, et al. (2022-08-21). "Toll-Like Receptor 4: A Promising Therapeutic Target for Alzheimer's Disease". Mediators of Inflammation. 2022: 7924199. doi: 10.1155/2022/7924199 . PMC   9420645 . PMID   36046763.
  96. 1 2 Leitner GR, Wenzel TJ, Marshall N, Gates EJ, Klegeris A (October 2019). "Targeting toll-like receptor 4 to modulate neuroinflammation in central nervous system disorders". Expert Opinion on Therapeutic Targets. 23 (10): 865–882. doi:10.1080/14728222.2019.1676416. PMID   31580163. S2CID   203652175.
  97. Michaud JP, Hallé M, Lampron A, Thériault P, Préfontaine P, Filali M, et al. (January 2013). "Toll-like receptor 4 stimulation with the detoxified ligand monophosphoryl lipid A improves Alzheimer's disease-related pathology". Proceedings of the National Academy of Sciences of the United States of America. 110 (5): 1941–1946. Bibcode:2013PNAS..110.1941M. doi: 10.1073/pnas.1215165110 . PMC   3562771 . PMID   23322736.
  98. Shavit Y, Wolf G, Goshen I, Livshits D, Yirmiya R (May 2005). "Interleukin-1 antagonizes morphine analgesia and underlies morphine tolerance". Pain. 115 (1–2): 50–59. doi:10.1016/j.pain.2005.02.003. PMID   15836969. S2CID   7286123.
  99. Mohan S, Davis RL, DeSilva U, Stevens CW (October 2010). "Dual regulation of mu opioid receptors in SK-N-SH neuroblastoma cells by morphine and interleukin-1β: evidence for opioid-immune crosstalk". Journal of Neuroimmunology. 227 (1–2): 26–34. doi:10.1016/j.jneuroim.2010.06.007. PMC   2942958 . PMID   20615556.
  100. Komatsu T, Sakurada S, Katsuyama S, Sanai K, Sakurada T (2009). Mechanism of allodynia evoked by intrathecal morphine-3-glucuronide in mice. International Review of Neurobiology. Vol. 85. pp. 207–19. doi:10.1016/S0074-7742(09)85016-2. ISBN   978-0-12-374893-5. PMID   19607972.
  101. Lewis SS, Hutchinson MR, Rezvani N, Loram LC, Zhang Y, Maier SF, et al. (January 2010). "Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via toll-like receptor 4/MD-2 and interleukin-1beta". Neuroscience. 165 (2): 569–583. doi:10.1016/j.neuroscience.2009.10.011. PMC   2795035 . PMID   19833175.
  102. Shen CH, Tsai RY, Shih MS, Lin SL, Tai YH, Chien CC, et al. (February 2011). "Etanercept restores the antinociceptive effect of morphine and suppresses spinal neuroinflammation in morphine-tolerant rats". Anesthesia and Analgesia. 112 (2): 454–459. doi: 10.1213/ANE.0b013e3182025b15 . PMID   21081778. S2CID   12295407.
  103. Hook MA, Washburn SN, Moreno G, Woller SA, Puga D, Lee KH, et al. (February 2011). "An IL-1 receptor antagonist blocks a morphine-induced attenuation of locomotor recovery after spinal cord injury". Brain, Behavior, and Immunity. 25 (2): 349–359. doi:10.1016/j.bbi.2010.10.018. PMC   3025088 . PMID   20974246.
  104. Watkins LR, Hutchinson MR, Rice KC, Maier SF (November 2009). "The "toll" of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia". Trends in Pharmacological Sciences. 30 (11): 581–591. doi:10.1016/j.tips.2009.08.002. PMC   2783351 . PMID   19762094.
  105. 1 2 3 Hutchinson MR, Zhang Y, Brown K, Coats BD, Shridhar M, Sholar PW, et al. (July 2008). "Non-stereoselective reversal of neuropathic pain by naloxone and naltrexone: involvement of toll-like receptor 4 (TLR4)". The European Journal of Neuroscience. 28 (1): 20–29. doi:10.1111/j.1460-9568.2008.06321.x. PMC   2588470 . PMID   18662331.
  106. Hutchinson MR, Coats BD, Lewis SS, Zhang Y, Sprunger DB, Rezvani N, et al. (November 2008). "Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia". Brain, Behavior, and Immunity. 22 (8): 1178–1189. doi:10.1016/j.bbi.2008.05.004. PMC   2783238 . PMID   18599265.
  107. Hutchinson MR, Lewis SS, Coats BD, Rezvani N, Zhang Y, Wieseler JL, et al. (May 2010). "Possible involvement of toll-like receptor 4/myeloid differentiation factor-2 activity of opioid inactive isomers causes spinal proinflammation and related behavioral consequences". Neuroscience. 167 (3): 880–893. doi:10.1016/j.neuroscience.2010.02.011. PMC   2854318 . PMID   20178837.
  108. Lin SL, Tsai RY, Tai YH, Cherng CH, Wu CT, Yeh CC, et al. (February 2010). "Ultra-low dose naloxone upregulates interleukin-10 expression and suppresses neuroinflammation in morphine-tolerant rat spinal cords". Behavioural Brain Research. 207 (1): 30–36. doi:10.1016/j.bbr.2009.09.034. PMID   19799935. S2CID   5128970.
  109. "Neuroscience: Making morphine work better". Nature. 484 (7395): 419. 26 April 2012. Bibcode:2012Natur.484Q.419.. doi: 10.1038/484419a . S2CID   52805136.
  110. Drahl C (22 August 2012). "Small Molecules Target Toll-Like Receptors". Chemical & Engineering News.
  111. Manček-Keber M, Jerala R (February 2015). "Postulates for validating TLR4 agonists". European Journal of Immunology. 45 (2): 356–370. doi:10.1002/eji.201444462. PMID   25476977. S2CID   32029412.
  112. Kim HM, Kim YM (October 2018). "HMGB1: LPS Delivery Vehicle for Caspase-11-Mediated Pyroptosis". Immunity. 49 (4): 582–584. doi: 10.1016/j.immuni.2018.09.021 . PMID   30332623.
  113. Romerio A, Peri F (2020). "Increasing the Chemical Variety of Small-Molecule-Based TLR4 Modulators: An Overview". Frontiers in Immunology. 11: 1210. doi: 10.3389/fimmu.2020.01210 . PMC   7381287 . PMID   32765484.
  114. 1 2 3 4 5 Hutchinson MR, Loram LC, Zhang Y, Shridhar M, Rezvani N, Berkelhammer D, et al. (June 2010). "Evidence that tricyclic small molecules may possess toll-like receptor and myeloid differentiation protein 2 activity". Neuroscience. 168 (2): 551–563. doi:10.1016/j.neuroscience.2010.03.067. PMC   2872682 . PMID   20381591.
  115. Chen F, Zou L, Williams B, Chao W (November 2021). "Targeting Toll-Like Receptors in Sepsis: From Bench to Clinical Trials". Antioxidants & Redox Signaling. 35 (15): 1324–1339. doi:10.1089/ars.2021.0005. PMC   8817700 . PMID   33588628.
  116. Jia ZJ, Wu FX, Huang QH, Liu JM (April 2012). "[Toll-like receptor 4: the potential therapeutic target for neuropathic pain]". Zhongguo Yi Xue Ke Xue Yuan Xue Bao. Acta Academiae Medicinae Sinicae. 34 (2): 168–173. doi:10.3881/j.issn.1000-503X.2012.02.013. PMID   22776604.
  117. Lan X, Han X, Li Q, Li Q, Gao Y, Cheng T, et al. (March 2017). "Pinocembrin protects hemorrhagic brain primarily by inhibiting toll-like receptor 4 and reducing M1 phenotype microglia". Brain, Behavior, and Immunity. 61: 326–339. doi:10.1016/j.bbi.2016.12.012. PMC   5453178 . PMID   28007523.
  118. Kaieda A, Takahashi M, Fukuda H, Okamoto R, Morimoto S, Gotoh M, et al. (December 2019). "Structure-Based Design, Synthesis, and Biological Evaluation of Imidazo[4,5-b]Pyridin-2-one-Based p38 MAP Kinase Inhibitors: Part 2". ChemMedChem. 14 (24): 2093–2101. doi:10.1002/cmdc.201900373. PMID   31697454. S2CID   207951964.
  119. 1 2 3 4 Hutchinson MR, Zhang Y, Shridhar M, Evans JH, Buchanan MM, Zhao TX, et al. (January 2010). "Evidence that opioids may have toll-like receptor 4 and MD-2 effects". Brain, Behavior, and Immunity. 24 (1): 83–95. doi:10.1016/j.bbi.2009.08.004. PMC   2788078 . PMID   19679181.
  120. Speer EM, Dowling DJ, Ozog LS, Xu J, Yang J, Kennady G, et al. (May 2017). "Pentoxifylline inhibits TLR- and inflammasome-mediated in vitro inflammatory cytokine production in human blood with greater efficacy and potency in newborns". Pediatric Research. 81 (5): 806–816. doi: 10.1038/pr.2017.6 . PMID   28072760. S2CID   47210724.
  121. Schüller SS, Wisgrill L, Herndl E, Spittler A, Förster-Waldl E, Sadeghi K, et al. (August 2017). "Pentoxifylline modulates LPS-induced hyperinflammation in monocytes of preterm infants in vitro". Pediatric Research. 82 (2): 215–225. doi: 10.1038/pr.2017.41 . PMID   28288151. S2CID   24897100.
  122. Neal MD, Jia H, Eyer B, Good M, Guerriero CJ, Sodhi CP, et al. (2013). "Discovery and validation of a new class of small molecule Toll-like receptor 4 (TLR4) inhibitors". PLOS ONE. 8 (6): e65779. Bibcode:2013PLoSO...865779N. doi: 10.1371/journal.pone.0065779 . PMC   3680486 . PMID   23776545.
  123. Impellizzeri D, Campolo M, Di Paola R, Bruschetta G, de Stefano D, Esposito E, et al. (2015). "Ultramicronized palmitoylethanolamide reduces inflammation an a Th1-mediated model of colitis". European Journal of Inflammation. 13: 14–31. doi: 10.1177/1721727X15575869 . S2CID   79398556.