Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 973
Filter
1.
Cell Commun Signal ; 22(1): 354, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972975

ABSTRACT

BACKGROUND: Hyperactive neutrophil extracellular traps (NETs) formation plays a crucial role in active severe systemic lupus erythematosus (SLE). However, what triggers the imbalance in dysregulated NETs formation in SLE is elusive. Transfer RNA-derived small RNAs (tsRNAs) are novel non-coding RNAs, which participate in various cellular processes. We explore the role of tsRNAs on NETs formation in SLE. METHODS: We analyzed the levels of NETs DNA and platelet-derived extracellular vesicles (pEVs) from 50 SLE patients and 20 healthy control subjects. The effects of pEVs on NETs formation were evaluated by using immunofluorescence assay and myeloperoxidase-DNA PicoGreen assay. The regulatory mechanism of pEVs on NETs formation and inflammatory cytokines production were investigated using an in vitro cell-based assay. RESULTS: Increased circulating NETs DNA and pEVs were shown in SLE patients and were associated with disease activity (P < 0.005). We demonstrated that SLE patient-derived immune complexes (ICs) induced platelet activation, followed by pEVs release. ICs-triggered NETs formation was significantly enhanced in the presence of pEVs through Toll-like receptor (TLR) 8 activation. Increased levels of tRF-His-GTG-1 in pEVs and neutrophils of SLE patients were associated with disease activity. tRF-His-GTG-1 interacted with TLR8 to prime p47phox phosphorylation in neutrophils, resulting in reactive oxygen species production and NETs formation. Additionally, tRF-His-GTG-1 modulated NF-κB and IRF7 activation in neutrophils upon TLR8 engagement, resulting IL-1ß, IL-8, and interferon-α upregulation, respectively. CONCLUSIONS: The level of tRF-His-GTG-1 was positively correlated with NETs formation in SLE patients; tRF-His-GTG-1 inhibitor could efficiently suppress ICs-triggered NETs formation/hyperactivation, which may become a potential therapeutic target.


Neutrophils and platelets are key members in the immunopathogenesis of SLE. EVs play a key role in intercellular communication. Abnormal NETs formation promotes vascular complications and organ damage in SLE patients. tsRNA is a novel regulatory small non-coding RNA and participates in diverse pathological processes. Herein, we showed that SLE patient-derived ICs activates platelets directly, followed by intracellular tRF-His-GTG-1 upregulation, which is loaded into pEVs. The pEV-carried tRF-His-GTG-1 could interact with TLR8 in neutrophils, followed by activation of the downstream signaling pathway, including p47phox-NOX2-ROS, which causes NETs enhancement, while IRF7 promotes the expression of IFN-α. The tRF-His-GTG-1 inhibitor could suppress efficiently SLE ICs-induced NETs formation and pEVs primed NETs enhancement. This study offers new molecular machinery to explain the association between the platelets-derived tsRNAs, pEVs, and hyperactive NETs formation in lupus. tRF-His-GTG-1 may serve as a potential therapeutic target and help to advance our understanding of tsRNAs in SLE pathogenesis.


Subject(s)
Extracellular Traps , Extracellular Vesicles , Interferon-alpha , Lupus Erythematosus, Systemic , Adult , Female , Humans , Male , Middle Aged , Blood Platelets/metabolism , Extracellular Traps/metabolism , Extracellular Vesicles/metabolism , Interferon-alpha/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/genetics , Neutrophils/metabolism , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics , RNA, Transfer/chemistry , RNA, Transfer/metabolism
2.
Sci Adv ; 10(27): eadg3747, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959314

ABSTRACT

Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.


Subject(s)
Adjuvants, Immunologic , Pyrimidines , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Humans , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/metabolism , Animals , Mice , Adjuvants, Immunologic/pharmacology , Toll-Like Receptor 7/agonists , Pyrimidines/pharmacology , Pyrimidines/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Imidazoles/pharmacology , Imidazoles/chemistry , THP-1 Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , COVID-19/virology , COVID-19/immunology , NF-kappa B/metabolism , Female , Drug Discovery/methods , Immunity, Innate/drug effects
3.
BMC Cancer ; 24(1): 824, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987740

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is ranked as the third most commonly diagnosed cancer and the third cause of cancer related deaths. CRC is greatly attributed to genetic and epigenetic mutations and immune dysregulation. Tumor aberrant expression of Toll-like Receptors (TLRs) can contribute to tumorigenesis. Recent studies suggested that microRNAs act as direct ligands of TLRs altering their expression and signaling pathways. AIM: To prove our concept that specific miRNA mimics may act as antagonists of their specific toll like receptors inhibiting their expression that could limit the release of pro-inflammatory and pro-tumorigenic cytokines leading to apoptosis of tumor cells. METHODS: From public microarray databases, we retrieved TLRs and miRNAs related to CRC followed by in silico docking of the selected miRNA ligands into the TLRs. Clinical validation after co-immunoprecipitation of TLRs and their interacting miRNA ligands was done. Expression of TLRs 1, 7,8 was determined by ELISA while miRNAs was measured by RT-qPCR. In addition, microRNA mimics of the down regulated miRNAs were transfected into human CRC cell lines. RESULTS: Our data demonstrate that TLRs 1, 7, 8 are up regulated in CRC compared to controls. Further, three miRNAs (-122, -29b and -15b) are relatively downregulated, while 4 miRNAs (-202, miRNA-98, -21 and -let7i) are upregulated in CRC patients compared to those with benign tumor and healthy controls. Transfection of down regulated miRNA mimics into CRC cell lines resulted in a significant reduction of the number and viability of cells as well as down regulating the expression of TLRs 1, 7 and 8 with ultimate reduction of downstream effector IL6 protein, suggesting that these miRNAs are negative regulators of carcinogenesis. CONCLUSION: MicroRNAs could act as antagonistic ligands of TLRs limiting the inflammatory tumor microenvironment.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , MicroRNAs , Toll-Like Receptor 8 , Tumor Microenvironment , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Tumor Microenvironment/genetics , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism , Cell Line, Tumor , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptors/metabolism , Toll-Like Receptors/genetics , Female , Male , Inflammation/genetics , Inflammation/metabolism , Signal Transduction
4.
Nat Commun ; 15(1): 5678, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971858

ABSTRACT

Inherited non-hemolytic anemia is a group of rare bone marrow disorders characterized by erythroid defects. Although concerted efforts have been made to explore the underlying pathogenetic mechanisms of these diseases, the understanding of the causative mutations are still incomplete. Here we identify in a diseased pedigree that a gain-of-function mutation in toll-like receptor 8 (TLR8) is implicated in inherited non-hemolytic anemia. TLR8 is expressed in erythroid lineage and erythropoiesis is impaired by TLR8 activation whereas enhanced by TLR8 inhibition from erythroid progenitor stage. Mechanistically, TLR8 activation blocks annexin A2 (ANXA2)-mediated plasma membrane localization of STAT5 and disrupts EPO signaling in HuDEP2 cells. TLR8 inhibition improves erythropoiesis in RPS19+/- HuDEP2 cells and CD34+ cells from healthy donors and inherited non-hemolytic anemic patients. Collectively, we identify a gene implicated in inherited anemia and a previously undescribed role for TLR8 in erythropoiesis, which could potentially be explored for therapeutic benefit in inherited anemia.


Subject(s)
Anemia , Erythropoiesis , Toll-Like Receptor 8 , Humans , Erythropoiesis/genetics , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics , Female , Anemia/genetics , Male , Pedigree , Erythropoietin/metabolism , Erythropoietin/genetics , Adult , Signal Transduction , Mutation , Erythroid Cells/metabolism , Animals , Erythroid Precursor Cells/metabolism
5.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38869500

ABSTRACT

UNC93B1 is a transmembrane domain protein mediating the signaling of endosomal Toll-like receptors (TLRs). We report five families harboring rare missense substitutions (I317M, G325C, L330R, R466S, and R525P) in UNC93B1 causing systemic lupus erythematosus (SLE) or chilblain lupus (CBL) as either autosomal dominant or autosomal recessive traits. As for a D34A mutation causing murine lupus, we recorded a gain of TLR7 and, to a lesser extent, TLR8 activity with the I317M (in vitro) and G325C (in vitro and ex vivo) variants in the context of SLE. Contrastingly, in three families segregating CBL, the L330R, R466S, and R525P variants were isomorphic with respect to TLR7 activity in vitro and, for R525P, ex vivo. Rather, these variants demonstrated a gain of TLR8 activity. We observed enhanced interaction of the G325C, L330R, and R466S variants with TLR8, but not the R525P substitution, indicating different disease mechanisms. Overall, these observations suggest that UNC93B1 mutations cause monogenic SLE or CBL due to differentially enhanced TLR7 and TLR8 signaling.


Subject(s)
Chilblains , Lupus Erythematosus, Systemic , Toll-Like Receptor 7 , Female , Humans , Male , Chilblains/genetics , Gain of Function Mutation , HEK293 Cells , Lupus Erythematosus, Cutaneous/genetics , Lupus Erythematosus, Cutaneous/pathology , Lupus Erythematosus, Systemic/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation, Missense , Pedigree , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism , Child, Preschool , Child , Young Adult , Adult
6.
ACS Biomater Sci Eng ; 10(7): 4374-4387, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38869358

ABSTRACT

DNA vaccines represent an innovative approach for the immunization of diverse diseases. However, their clinical trial outcomes are constrained by suboptimal transfection efficiency and immunogenicity. In this work, we present a universal methodology involving the codelivery of Toll-like receptor 7/8 agonists (TLR7/8a) and antigen gene using TLR7/8a-conjugated peptide-coated poly(ß-amino ester) (PBAE) nanoparticles (NPs) to augment delivery efficiency and immune response. Peptide-TLR7/8a-coated PBAE NPs exhibit advantageous biophysical attributes, encompassing diminutive particle dimensions, nearly neutral ζ potential, and stability in the physiological environment. This synergistic approach not only ameliorates the stability of plasmid DNA (pDNA) and gene delivery efficacy but also facilitates subsequent antigen production. Furthermore, under optimal formulation conditions, the TLR7/8a-conjugated peptide coated PBAE NPs exhibit a potent capacity to induce robust immune responses. Collectively, this nanoparticulate gene delivery system demonstrates heightened transfection efficacy, stability, biodegradability, immunostimulatory effect, and low toxicity, making it a promising platform for the clinical advancement of DNA vaccines.


Subject(s)
Nanoparticles , Peptides , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Vaccines, DNA , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Toll-Like Receptor 8/immunology , Toll-Like Receptor 8/agonists , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/immunology , Animals , Nanoparticles/chemistry , Peptides/chemistry , Peptides/immunology , Humans , Mice , Female , Polymers/chemistry , Plasmids/genetics , Plasmids/immunology , Mice, Inbred C57BL
7.
Int J Biol Macromol ; 270(Pt 1): 132258, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735613

ABSTRACT

Covalently linking an adjuvant to an antigenic protein enhances its immunogenicity by ensuring a synergistic delivery to the immune system, fostering a more robust and targeted immune response. Most adjuvant-protein conjugate vaccines incorporate only one adjuvant due to the difficulties in its synthesis. However, there is a growing interest in developing vaccines with multiple adjuvants designed to elicit a more robust and targeted immune response by engaging different aspects of the immune system for complex diseases where traditional vaccines fall short. Here, we pioneer the synthesis of a dual-adjuvants protein conjugate Vaccine 1 by assembling a toll-like receptor 7/8 (TLR7/8) agonist, an invariant natural killer T cell (iNKT) agonist with a clickable bicyclononyne (BCN). The BCN group can bio-orthogonally react with azide-modified severe acute respiratory syndrome coronavirus-2 receptor-binding domain (SARS-CoV-2 RBD) trimer antigen to give the three-component Vaccine 1. Notably, with a mere 3 µg antigen, it elicited a balanced subclass of IgG titers and 20-fold more IgG2a than control vaccines, highlighting its potential for enhancing antibody-dependent cellular cytotoxicity. This strategy provides a practicable way to synthesize covalently linked dual immunostimulants. It expands the fully synthetic self-adjuvant protein vaccine that uses a single adjuvant to include two different types of adjuvants.


Subject(s)
Adjuvants, Immunologic , COVID-19 Vaccines , COVID-19 , Natural Killer T-Cells , SARS-CoV-2 , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/immunology , SARS-CoV-2/immunology , Animals , Natural Killer T-Cells/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/pharmacology , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/immunology , Humans , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Mice , COVID-19/prevention & control , COVID-19/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/immunology , Female , Adjuvants, Vaccine/chemistry , Adjuvants, Vaccine/pharmacology , Immunoglobulin G/immunology
8.
J Nanobiotechnology ; 22(1): 296, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811964

ABSTRACT

BACKGROUND: Combination therapy involving immune checkpoint blockade (ICB) and other drugs is a potential strategy for converting immune-cold tumors into immune-hot tumors to benefit from immunotherapy. To achieve drug synergy, we developed a homologous cancer cell membrane vesicle (CM)-coated metal-organic framework (MOF) nanodelivery platform for the codelivery of a TLR7/8 agonist with an epigenetic inhibitor. METHODS: A novel biomimetic codelivery system (MCM@UN) was constructed by MOF nanoparticles UiO-66 loading with a bromodomain-containing protein 4 (BRD4) inhibitor and then coated with the membrane vesicles of homologous cancer cells that embedding the 18 C lipid tail of 3M-052 (M). The antitumor immune ability and tumor suppressive effect of MCM@UN were evaluated in a mouse model of triple-negative breast cancer (TNBC) and in vitro. The tumor immune microenvironment was analyzed by multicolor immunofluorescence staining. RESULTS: In vitro and in vivo data showed that MCM@UN specifically targeted to TNBC cells and was superior to the free drug in terms of tumor growth inhibition and antitumor immune activity. In terms of mechanism, MCM@UN blocked BRD4 and PD-L1 to prompt dying tumor cells to disintegrate and expose tumor antigens. The disintegrated tumor cells released damage-associated molecular patterns (DAMPs), recruited dendritic cells (DCs) to efficiently activate CD8+ T cells to mediate effective and long-lasting antitumor immunity. In addition, TLR7/8 agonist on MCM@UN enhanced lymphocytes infiltration and immunogenic cell death and decreased regulatory T-cells (Tregs). On clinical specimens, we found that mature DCs infiltrating tumor tissues of TNBC patients were negatively correlated with the expression of BRD4, which was consistent with the result in animal model. CONCLUSION: MCM@UN specifically targeted to TNBC cells and remodeled tumor immune microenvironment to inhibit malignant behaviors of TNBC.


Subject(s)
Toll-Like Receptor 7 , Toll-Like Receptor 8 , Triple Negative Breast Neoplasms , Tumor Microenvironment , Animals , Triple Negative Breast Neoplasms/drug therapy , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Mice , Female , Humans , Cell Line, Tumor , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Transcription Factors/metabolism , Mice, Inbred BALB C , Cell Cycle Proteins/metabolism , Immunotherapy/methods , Epigenesis, Genetic/drug effects , Bromodomain Containing Proteins
9.
Int J Biol Macromol ; 269(Pt 1): 132018, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38702002

ABSTRACT

Toll-like receptor 8 (TLR8), an important innate immune receptor recognizing single stranded RNA and the antiviral imidazoquinoline compounds, can activate intracellular signaling pathway and produce an inflammatory response to kill and eliminate pathogens. However, the molecular regulation mechanisms of TLR8 signaling and its anti-infection activity are not fully elucidated. Our previous transcriptome analysis of porcine TLR8 (pTLR8) signaling suggested the immune checkpoint receptor TIM-3 as the potential regulator for pTLR8. Here we investigated TIM-3 in the regulation of pTLR8 signaling and its anti-infection activity. Our results showed that porcine TIM-3 is upregulated by pTLR8 signaling and TIM-3 inhibits pTLR8 signaling activity in a negative feedback way. Accordingly, TIM-3 disturbs pTLR8 mediated anti-bacterial and anti-viral activity. Mechanistically, TIM-3 suppresses PI3K-AKT pathway by inhibiting the TLR8-PI3K p85 interaction and subsequent AKT phosphorylation which is essential for TLR8 signaling and anti-infection activity. Therefore, our study reveals new insights into innate immune TLR8 signaling and its anti-infection function.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Toll-Like Receptor 8 , Animals , Humans , Hepatitis A Virus Cellular Receptor 2/metabolism , Immunity, Innate/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Swine , Toll-Like Receptor 8/metabolism , HEK293 Cells , Vero Cells
10.
PLoS One ; 19(5): e0300778, 2024.
Article in English | MEDLINE | ID: mdl-38758816

ABSTRACT

Mpox (formerly known as monkeypox) virus and some related poxviruses including smallpox virus pose a significant threat to public health, and effective prevention and treatment strategies are needed. This study utilized a reverse vaccinology approach to retrieve conserved epitopes for monkeypox virus and construct a vaccine that could provide cross-protection against related viruses with similar antigenic properties. The selected virulent proteins of monkeypox virus, MPXVgp165, and Virion core protein P4a, were subjected to epitope mapping for vaccine construction. Two vaccines were constructed using selected T cell epitopes and B cell epitopes with PADRE and human beta-defensins adjuvants conjugated in the vaccine sequence. Both constructs were found to be highly antigenic, non-allergenic, nontoxic, and soluble, suggesting their potential to generate an adequate immune response and be safe for humans. Vaccine construct 1 was selected for molecular dynamic simulation studies. The simulation studies revealed that the TLR8-vaccine complex was more stable than the TLR3-vaccine complex. The lower RMSD and RMSF values of the TLR8 bound vaccine compared to the TLR3 bound vaccine suggested better stability and consistency of hydrogen bonds. The Rg values of the vaccine chain bound to TLR8 indicated overall stability, whereas the vaccine chain bound to TLR3 showed deviations throughout the simulation. These results suggest that the constructed vaccine could be a potential preventive measure against monkeypox and related viruses however, further experimental validation is required to confirm these findings.


Subject(s)
Molecular Dynamics Simulation , Monkeypox virus , Humans , Monkeypox virus/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Computer Simulation , Poxviridae/immunology , Viral Vaccines/immunology , Epitope Mapping , Mpox (monkeypox)/prevention & control , Mpox (monkeypox)/immunology , Animals , Toll-Like Receptor 8/immunology
11.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791389

ABSTRACT

The pathogenesis of systemic lupus erythematosus (SLE) is linked to the differential roles of toll-like receptors (TLRs), particularly TLR7, TLR8, and TLR9. TLR7 overexpression or gene duplication, as seen with the Y-linked autoimmune accelerator (Yaa) locus or TLR7 agonist imiquimod, correlates with increased SLE severity, and specific TLR7 polymorphisms and gain-of-function variants are associated with enhanced SLE susceptibility and severity. In addition, the X-chromosome location of TLR7 and its escape from X-chromosome inactivation provide a genetic basis for female predominance in SLE. The absence of TLR8 and TLR9 have been shown to exacerbate the detrimental effects of TLR7, leading to upregulated TLR7 activity and increased disease severity in mouse models of SLE. The regulatory functions of TLR8 and TLR9 have been proposed to involve competition for the endosomal trafficking chaperone UNC93B1. However, recent evidence implies more direct, regulatory functions of TLR9 on TLR7 activity. The association between age-associated B cells (ABCs) and autoantibody production positions these cells as potential targets for treatment in SLE, but the lack of specific markers necessitates further research for precise therapeutic intervention. Therapeutically, targeting TLRs is a promising strategy for SLE treatment, with drugs like hydroxychloroquine already in clinical use.


Subject(s)
Lupus Erythematosus, Systemic , Toll-Like Receptor 7 , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Animals , Humans , Mice , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptors/metabolism , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics , Disease Models, Animal , Genetic Predisposition to Disease
12.
Chem Commun (Camb) ; 60(42): 5474-5485, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38712400

ABSTRACT

Toll-like receptor 7/8 (TLR-7/8) agonists serve as a promising class of pattern recognition receptors that effectively evoke the innate immune response, making them promising immunomodulatory agents for tumor immunotherapy. However, the uncontrollable administration of TLR-7/8 agonists frequently leads to the occurrence of severe immune-related adverse events (irAEs). Thus, it is imperative to strategically design tumor-microenvironment-associated biomarkers or exogenous stimuli responsive TLR-7/8 agonists in order to accurately evaluate and activate innate immune responses. No comprehensive elucidation has been documented thus far regarding TLR-7/8 immune agonists that are specifically engineered to enhance immune activation. In this feature article, we provide an overview of the advancements in TLR-7/8 agonists, aiming to enhance the comprehension of their mechanisms and promote the clinical progression through nanomedicine strategies. The current challenges and future directions of cancer immunotherapy are also discussed, with the hope that this work will inspire researchers to explore innovative applications for triggering immune responses through TLR-7/8 agonists.


Subject(s)
Toll-Like Receptor 7 , Toll-Like Receptor 8 , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Humans , Immunotherapy , Neoplasms/drug therapy , Neoplasms/immunology , Immunity, Innate/drug effects , Animals
13.
Immunology ; 172(4): 577-587, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38631842

ABSTRACT

Preterm birth is the largest contributor to neonatal morbidity and is often associated with chorioamnionitis, defined as inflammation/infection of the fetal membranes (FMs). Chorioamnionitis is characterised by neutrophil infiltration of the FMs and is associated with elevated levels of the neutrophil chemoattractant, interleukin (IL)-8 and the proinflammatory cytokine, IL-1ß. While FMs can respond to infections through innate immune sensors, such as toll-like receptors (TLRs), the downstream mechanisms by which chorioamnionitis arises are not fully understood. A novel group of non-classical microRNAs (miR-21a, miR-29a, miR-146a-3p, Let-7b) function as endogenous danger signals by activating the ssRNA viral sensors TLR7 and TLR8. In this study, the pro-inflammatory roles of TLR7/TLR8-activating miRs were examined as mediators of FM inflammation in response to bacterial lipopolysaccharide (LPS) using an in vitro human FM explant system, an in vivo mouse model of pregnancy, and human clinical samples. Following LPS exposure, miR-146a-3p was significantly increased in both human FM explants and wild-type mouse FMs. Expression of miR-146a-3p was also significantly elevated in FMs from women with preterm birth and chorioamnionitis. FM IL-8 and inflammasome-mediated IL-1ß production in response to LPS was dependent on miR-146a-3p and TLR8 downstream of TLR4 activation. In wild-type mice, LPS exposure increased FM IL-8 and IL-1ß production and induced preterm birth. In TLR7-/-/TLR8-/- mice, LPS exposure was able to initiate but not sustain preterm birth, and FM inflammation was reduced. Together, we demonstrate a novel signalling mechanism at the maternal-fetal interface in which TLR8-activating miR-146a-3p acts as an intermediate danger signal to drive FM inflammasome-dependent and -independent mechanisms of inflammation and, thus, may play a role in chorioamnionitis and subsequent preterm birth.


Subject(s)
Chorioamnionitis , Extraembryonic Membranes , Lipopolysaccharides , MicroRNAs , Toll-Like Receptor 8 , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Pregnancy , Female , Chorioamnionitis/immunology , Chorioamnionitis/metabolism , Humans , Extraembryonic Membranes/metabolism , Extraembryonic Membranes/immunology , Mice , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics , Signal Transduction , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Mice, Knockout , Inflammation/immunology , Inflammation/metabolism , Mice, Inbred C57BL , Interleukin-1beta/metabolism , Premature Birth/immunology , Interleukin-8/metabolism
14.
Neuromolecular Med ; 26(1): 16, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38668900

ABSTRACT

Toll-like receptor (TLR) 7 plays an important role in recognizing virus-derived nucleic acids. TLR7 signaling in astrocytes and microglia is critical for activating immune responses against neurotrophic viruses. Neurons express TLR7, similar to glial cells; however, the role of neuronal TLR7 has not yet been fully elucidated. This study sought to determine whether resiquimod, the TLR7/8 agonist, induces the expression of inflammatory chemokines in SH-SY5Y human neuroblastoma cells. Immunofluorescence microscopy revealed that TLR7 was constitutively expressed in SH-SY5Y cells. Stimulation with resiquimod induced C-C motif chemokine ligand 2 (CCL2) expression, accompanied by the activation of nuclear factor-kappa B (NF-κB) in SH-SY5Y cells. Resiquimod increased mRNA levels of C-X-C motif chemokine ligand 8 (CXCL8) and CXCL10, while the increase was slight at the protein level. Knockdown of NF-κB p65 eliminated resiquimod-induced CCL2 production. This study provides novel evidence that resiquimod has promising therapeutic potential against central nervous system viral infections through its immunostimulatory effects on neurons.


Subject(s)
Chemokine CCL2 , Chemokine CXCL10 , Imidazoles , Interleukin-8 , Toll-Like Receptor 7 , Transcription Factor RelA , Humans , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL2/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL10/biosynthesis , Imidazoles/pharmacology , Interleukin-8/genetics , Interleukin-8/biosynthesis , Neuroblastoma , Neurons/drug effects , Neurons/metabolism , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/genetics , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics
15.
Immun Inflamm Dis ; 12(4): e1211, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38602270

ABSTRACT

BACKGROUND: Traumatic cartilage injury is an important cause of osteoarthritis (OA) and limb disability, and toll-like receptors (TLRs) mediated innate immune response has been confirmed to play a crucial role in cartilage injury. In the previous study, we found that the activation of TLR8 molecules in injured articular cartilage was more obvious than other TLRs by establishing an animal model of knee impact injury in rabbits, and the changes of TLR8 molecules could significantly affect the process of articular cartilage injury and repair. OBJECTIVE: To verify how mir-99a-5p regulates TLR8 receptor mediated innate immune response to treat traumatic cartilage injury. METHODS: The impact of a heavy object on the medial condyle of the rabbit's knee joint caused damage to the medial condylar cartilage. Through pathological and imaging analysis, it was demonstrated whether the establishment of an animal model of traumatic cartilage injury was successful. Establishing a cell model by virus transfection of chondrocytes to demonstrate the role of TLR8 in the innate immune response to impact cartilage injury. Through transcriptome sequencing, potential targets of TLR8, mir-99a-5p, were predicted, and basic experiments were conducted to demonstrate how they interact with innate immune responses to impact cartilage damage. RESULTS: TLR8 is a receptor protein of the immune system, which is widely expressed in immune cells. In our study, we found that TLR8 expression is localized in lysosomes and endosomes. Mir-99a-5p can negatively regulate TLR8 to activate PI3K-AKT molecular pathway and aggravate cartilage damage. Inhibiting TLR8 expression can effectively reduce the incidence of articular cartilage damage. CONCLUSION: Based on the results from this study, mir-99a-5p may be an effective molecular marker for predicting traumatic cartilage injury and targeting TLR8 is a novel and promising approach for the prevention or early treatment of cartilage damage.


Subject(s)
Cartilage, Articular , MicroRNAs , Animals , Rabbits , MicroRNAs/genetics , Toll-Like Receptor 8/metabolism , Phosphatidylinositol 3-Kinases , Knee Joint/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology
16.
Biochem Biophys Res Commun ; 709: 149790, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38564938

ABSTRACT

Macrophages play an important role in the development and progression of acute rejection after kidney transplantation. The study aims to investigate the biological role and significance of macrophage-associated genes (MAG) in acute rejection after kidney transplantation. We utilized transcriptome sequencing results from public databases related to acute rejection of kidney transplantation for comprehensive analysis and validation in animal experiments. We found that a large number of immune-related signaling pathways are activated in acute rejection. PPI protein interaction networks and machine learning were used to establish a Hub gene consisting of TYROBP and TLR8 for the diagnosis of acute rejection. The single-gene GSEA enrichment analysis and immune cell correlation analysis revealed a close correlation between the expression of Hub genes and immune-related biological pathways as well as the expression of multiple immune cells. In addition, the study of TF, miRNAs, and drugs provided a theoretical basis for regulating and treating the Hub genes in acute rejection. Finally, the animal experiments demonstrated once again that acute rejection can aggravate kidney tissue damage, apoptosis level, and increase the release of inflammatory factors. We established and validated a macrophage-associated diagnostic model for acute rejection after kidney transplantation, which can accurately diagnose the biological alterations in acute rejection after kidney transplantation.


Subject(s)
Kidney Transplantation , Animals , Kidney Transplantation/adverse effects , Toll-Like Receptor 8 , Gene Expression Profiling , Biomarkers , Macrophages
17.
Cell Commun Signal ; 22(1): 220, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589923

ABSTRACT

Endosomal single-stranded RNA-sensing Toll-like receptor-7/8 (TLR7/8) plays a pivotal role in inflammation and immune responses and autoimmune diseases. However, the mechanisms underlying the initiation of the TLR7/8-mediated autoimmune signaling remain to be fully elucidated. Here, we demonstrate that miR-574-5p is aberrantly upregulated in tissues of lupus prone mice and in the plasma of lupus patients, with its expression levels correlating with the disease activity. miR-574-5p binds to and activates human hTLR8 or its murine ortholog mTlr7 to elicit a series of MyD88-dependent immune and inflammatory responses. These responses include the overproduction of cytokines and interferons, the activation of STAT1 signaling and B lymphocytes, and the production of autoantigens. In a transgenic mouse model, the induction of miR-574-5p overexpression is associated with increased secretion of antinuclear and anti-dsDNA antibodies, increased IgG and C3 deposit in the kidney, elevated expression of inflammatory genes in the spleen. In lupus-prone mice, lentivirus-mediated silencing of miR-574-5p significantly ameliorates major symptoms associated with lupus and lupus nephritis. Collectively, these results suggest that the miR-574-5p-hTLR8/mTlr7 signaling is an important axis of immune and inflammatory responses, contributing significantly to the development of lupus and lupus nephritis.


Subject(s)
Lupus Nephritis , MicroRNAs , Humans , Mice , Animals , Lupus Nephritis/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism , Kidney/metabolism , Mice, Transgenic , MicroRNAs/genetics
18.
Adv Mater ; 36(23): e2311574, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38433564

ABSTRACT

Dendritic cell (DC) maturation is a crucial process for antigen presentation and the initiation of T cell-mediated immune responses. Toll-like receptors play pivotal roles in stimulating DC maturation and promoting antigen presentation. Here, a novel message RNA (mRNA) cancer vaccine is reported that boosts antitumor efficacy by codelivering an mRNA encoding tumor antigen and a TLR7/8 agonist (R848) to DC using supramolecular lipid nanoparticles (SMLNP) as a delivery platform, in which a new ionizable lipid (N2-3L) remarkably enhances the translation efficiency of mRNA and a ß-cyclodextrin (ß-CD)-modified ionizable lipid (Lip-CD) encapsulates R848. The incorporation of R848 adjuvant into the mRNA vaccine through noncovalent host-guest complexation significantly promotes DC maturation and antigen presentation after vaccination, thus resulting in superior antitumor efficacy in vivo. Moreover, the antitumor efficacy is further boosted synergized with immune checkpoint blockade by potentiating the anticancer capability of cytotoxic T lymphocytes infiltrated in tumor sites. This work indicates that SMLNP shows brilliant potential as next-generation delivery system in the development of mRNA vaccines with high efficacy.


Subject(s)
Cancer Vaccines , Dendritic Cells , Imidazoles , Immunotherapy , Lipids , Nanoparticles , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Animals , Nanoparticles/chemistry , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Dendritic Cells/immunology , Mice , Lipids/chemistry , Imidazoles/chemistry , mRNA Vaccines/chemistry , beta-Cyclodextrins/chemistry , RNA, Messenger/genetics , RNA, Messenger/chemistry , Neoplasms/therapy , Cell Line, Tumor , Antigens, Neoplasm/immunology , Humans , Mice, Inbred C57BL , Liposomes
19.
J Autoimmun ; 145: 103189, 2024 May.
Article in English | MEDLINE | ID: mdl-38442677

ABSTRACT

OBJECTIVES: Monocyte-derived dendritic cells (DCs) are key players in the induction of inflammation, autoreactive T cell activation and loss of tolerance in rheumatoid arthritis (RA), but the precise mechanisms underlying their activation remain elusive. Here, we hypothesized that extracellular microRNAs released in RA synovial fluids may represent a novel, physiological stimulus triggering unwanted immune response via TLR8-expressing DC stimulation. METHODS: Human monocyte-derived DCs were stimulated with a mixture of GU-rich miRNAs upregulated in RA tissues and released in synovial fluids (Ex-miRNAs). Activation of DCs was assessed in terms of NF-κB activation by Western blot, cytokine production by ELISA, T cell proliferation and polarization by allogeneic mixed lymphocyte reaction. DC differentiation into osteoclasts was evaluated in terms of tartrate-resistant acid phosphatase production and formation of resorption pits in dentine slices. Induction of joint inflammation in vivo was evaluated using a murine model of DC-induced arthritis. TLR7/8 involvement was assessed by specific inhibitors. RESULTS: Ex-miRNAs activate DCs to secrete TNFα, induce joint inflammation, start an early autoimmune response and potentiate the differentiation of DCs into aggressive osteoclasts. CONCLUSIONS: This work represents a proof of concept that the pool of extracellular miRNAs overexpressed in RA joints can act as a physiological activator of inflammation via the stimulation of TLR8 expressed by human DCs, which in turn exert arthritogenic functions. In this scenario, pharmacological inhibition of TLR8 might offer a new therapeutic option to reduce inflammation and osteoclast-mediated bone destruction in RA.


Subject(s)
Arthritis, Rheumatoid , Cell Differentiation , Dendritic Cells , MicroRNAs , Osteoclasts , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , MicroRNAs/genetics , Toll-Like Receptor 8/metabolism , Osteoclasts/metabolism , Osteoclasts/immunology , Animals , Toll-Like Receptor 7/metabolism , Mice , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Synovial Fluid/immunology , Synovial Fluid/metabolism , Cells, Cultured , Female , Male
20.
Virol J ; 21(1): 67, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38509569

ABSTRACT

Since 1997, highly pathogenic avian influenza viruses, such as H5N1, have been recognized as a possible pandemic hazard to men and the poultry business. The rapid rate of mutation of H5N1 viruses makes the whole process of designing vaccines extremely challenging. Here, we used an in silico approach to design a multi-epitope vaccine against H5N1 influenza A virus using hemagglutinin (HA) and neuraminidase (NA) antigens. B-cell epitopes, Cytotoxic T lymphocyte (CTL) and Helper T lymphocyte (HTL) were predicted via IEDB, NetMHC-4 and NetMHCII-2.3 respectively. Two adjuvants consisting of Human ß-defensin-3 (HßD-3) along with pan HLA DR-binding epitope (PADRE) have been chosen to induce more immune response. Linkers including KK, AAY, HEYGAEALERAG, GPGPGPG and double EAAAK were utilized to link epitopes and adjuvants. This construct encodes a protein having 350 amino acids and 38.46 kDa molecular weight. Antigenicity of ~ 1, the allergenicity of non-allergen, toxicity of negative and solubility of appropriate were confirmed through Vaxigen, AllerTOP, ToxDL and DeepSoluE, respectively. The 3D structure of H5N1 was refined and validated with a Z-Score of - 0.87 and an overall Ramachandran of 99.7%. Docking analysis showed H5N1 could interact with TLR7 (docking score of - 374.08 and by 4 hydrogen bonds) and TLR8 (docking score of - 414.39 and by 3 hydrogen bonds). Molecular dynamics simulations results showed RMSD and RMSF of 0.25 nm and 0.2 for H5N1-TLR7 as well as RMSD and RMSF of 0.45 nm and 0.4 for H5N1-TLR8 complexes, respectively. Molecular Mechanics Poisson-Boltzmann Surface Area (MM/PBSA) confirmed stability and continuity of interaction between H5N1-TLR7 with the total binding energy of - 29.97 kJ/mol and H5N1-TLR8 with the total binding energy of - 23.9 kJ/mol. Investigating immune response simulation predicted evidence of the ability to stimulate T and B cells of the immunity system that shows the merits of this H5N1 vaccine proposed candidate for clinical trials.


Subject(s)
Influenza A Virus, H5N1 Subtype , Vaccines , Animals , Humans , Influenza A Virus, H5N1 Subtype/genetics , Epitopes, T-Lymphocyte/genetics , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Epitopes, B-Lymphocyte , Computational Biology/methods , Molecular Docking Simulation , Vaccines, Subunit/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...