Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 317
Filter
1.
Viruses ; 16(4)2024 04 17.
Article in English | MEDLINE | ID: mdl-38675965

ABSTRACT

Epstein-Barr virus (EBV), a Herpesviridae family member, is associated with an increased risk of autoimmune disease development in the host. We previously demonstrated that EBV DNA elevates levels of the pro-inflammatory cytokine IL-17A and that inhibiting Toll-like receptor (TLR) 3, 7, or 9 reduces its levels. Moreover, this DNA exacerbated colitis in a mouse model of inflammatory bowel disease (IBD). In the study at hand, we examined whether inhibition of TLR3, 7, or 9 alleviates this exacerbation. Mice were fed 1.5% dextran sulfate sodium (DSS) water and administered EBV DNA. Then, they were treated with a TLR3, 7, or 9 inhibitor or left untreated. We also assessed the additive impact of combined inhibition of all three receptors. Mice that received DSS, EBV DNA, and each inhibitor alone, or a combination of inhibitors, showed significant improvement. They also had a decrease in the numbers of the pathogenic colonic IL-17A+IFN-γ+ foci. Inhibition of all three endosomal TLR receptors offered no additive benefit over administering a single inhibitor. Therefore, inhibition of endosomal TLRs reduces EBV DNA exacerbation of mouse colitis, offering a potential approach for managing IBD patients infected with EBV.


Subject(s)
DNA, Viral , Herpesvirus 4, Human , Inflammatory Bowel Diseases , Toll-Like Receptors , Animals , Female , Mice , Colitis/chemically induced , Colitis/drug therapy , Colitis/virology , Dextran Sulfate , Disease Models, Animal , DNA, Viral/adverse effects , DNA, Viral/pharmacology , Endosomes/drug effects , Endosomes/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/drug therapy , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/virology , Interleukin-17/metabolism , Mice, Inbred C57BL , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/metabolism , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/metabolism
2.
Clin Pharmacol Ther ; 115(6): 1346-1357, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38415785

ABSTRACT

Enpatoran is a novel, highly selective, and potent dual toll-like receptor (TLR)7 and TLR8 inhibitor currently under development for the treatment of autoimmune disorders including systemic lupus erythematosus (SLE), cutaneous lupus erythematosus (CLE), and myositis. The ongoing phase II study (WILLOW; NCT05162586) is evaluating enpatoran for 24 weeks in patients with active SLE or CLE and is currently recruiting. To support development of WILLOW as an Asia-inclusive multiregional clinical trial (MRCT) according to International Conference on Harmonisation E5 and E17 principles, we have evaluated ethnic sensitivity to enpatoran based on clinical pharmacokinetic (PK), pharmacodynamic (PD), and safety data from an ethno-bridging study (NCT04880213), supplemented by relevant quantitative PK, PD, and disease trajectory modeling (DTM) results, and drug metabolism/disease knowledge. A single-center, open-label, sequential dose group study in White and Japanese subjects matched by body weight, height, and sex demonstrated comparable PK and PD properties for enpatoran in Asian vs. non-Asian (White and other) subjects across single 100, 200, and 300 mg orally administered doses. DTM suggested no significant differences in SLE disease trajectory for Asian vs. non-Asian individuals. Aldehyde oxidase (AOX) is considered to be a key contributor to enpatoran metabolism, and a literature review indicated no relevant ethnic differences in AOX function based on in vitro and clinical PK data from marketed drugs metabolized by AOX, supporting the conclusion of low ethnic sensitivity for enpatoran. Taken together, the inclusion of Asian patients in MRCTs including WILLOW was informed based on a Totality of Evidence approach.


Subject(s)
Lupus Erythematosus, Systemic , Toll-Like Receptors , Adult , Female , Humans , Male , Middle Aged , Asia , Lupus Erythematosus, Cutaneous/drug therapy , Lupus Erythematosus, Systemic/drug therapy , Research Design , Clinical Trials, Phase II as Topic , Toll-Like Receptors/antagonists & inhibitors , East Asian People , White
3.
Chem Biodivers ; 20(4): e202300025, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36898972

ABSTRACT

Toll-like receptors (TLRs) recognize pathogen-associated molecular patterns and trigger an inflammatory response via the myeloid differential factor 88 (MyD88)-dependent and toll-interleukin-1 receptor domain-containing adapter-inducing interferon-ß (TRIF)-dependent pathways. Lindenane type sesquiterpene dimers (LSDs) are characteristic metabolites of plants belonging to the genus Sarcandra (Chloranthaceae). The aim of this study was to evaluate the potential anti-inflammatory effects of the LSDs shizukaol D (1) and sarcandrolide E (2) on lipopolysaccharides (LPS)-stimulated RAW264.7 macrophages in vitro, and explore the underlying mechanisms. Both LSDs neutralized the LPS-induced morphological changes and production of nitric oxide (NO), as determined by CCK-8 assay and Griess assay, respectively. Furthermore, shizukaol D (1) and sarcandrolide E (2) downregulated interferon ß (IFNß), tumor necrosis factor α (TNFα) and interleukin-1ß (IL-1ß) mRNA levels as measured by reverse transcription polymerase chain reaction (RT-PCR), and inhibited the phosphorylation of nuclear factor kappa B p65 (p65), nuclear factor kappa-Bα (IκBα), Jun N-terminal kinase (JNK), extracellular regulated kinase (ERK), mitogen-activated protein kinase p38 (p38), MyD88, IL-1RI-associated protein kinase 1 (IRAK1), and transforming growth factor-ß-activated kinase 1 (TAK1) proteins in the Western blotting assay. In conclusion, LSDs can alleviate the inflammatory response by inhibiting the TLR/MyD88 signalling pathway.


Subject(s)
Inflammation , Sesquiterpenes , Toll-Like Receptors , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Sesquiterpenes/pharmacology , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/metabolism
4.
Biomed Pharmacother ; 147: 112622, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35008000

ABSTRACT

Nucleic acid-mediated interferon signaling plays a pivotal role in defense against microorganisms, especially during viral infection. Receptors sensing exogenous nucleic acid molecules are localized in the cytosol and endosomes. Cytosolic sensors, including cGAS, RIG-I, and MDA5, and endosome-anchored receptors are toll-like receptors (TLR3, TLR7, TLR8, and TLR9). These TLRs share the same domain architecture and have similar structures, facing the interior of endosomes so their binding to nucleic acids of invading pathogens via endocytosis is possible. The correct function of these receptors is crucial for cell homeostasis and effective response against pathogen invasion. A variety of endogenous mechanisms modulates their activities. Nevertheless, naturally occurring mutations lead to aberrant TLR-mediated interferon (IFN) signaling. Furthermore, certain pathogens require a more robust defense against control. Thus, manipulating these TLR activities has a profound impact. High-throughput virtual screening followed by experimental validation led to the discovery of numerous chemicals that can change these TLR-mediated IFN signaling activities. Many of them are unique in selectivity, while others regulate more than one TLR due to commonalities in these receptors. We summarized these nucleic acid-sensing TLR-mediated IFN signaling pathways and the corresponding chemicals activating or deactivating their signaling.


Subject(s)
Antiviral Agents/pharmacology , Interferons/metabolism , Nucleic Acids/pharmacology , Toll-Like Receptors/drug effects , Antiviral Agents/chemistry , Humans , Immunity, Innate , Nucleic Acids/chemistry , Toll-Like Receptors/agonists , Toll-Like Receptors/antagonists & inhibitors
5.
Acta Biomater ; 141: 132-139, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35032721

ABSTRACT

The clinical success of Toll-like receptor (TLR) agonists is based on their capacity to efficiently mobilize both innate and adaptive immunity. However, rapid distribution of TLR agonists into the systemic circulation may result in systemic cytokine storms. Telratolimod (Tel) is a TLR 7/8 agonist whose structure has a hydrophobic long chain that helps to prolong its release. Despite this, the phase I study of Tel showed cytokine release syndromes in 3/35 patients. Herein, we designed an injectable phase transition gel (PGE) that served as a superior drug depot for fatty acid-modified drugs. PGE further minimized the systemic drug exposure of Tel and the possible cytokine storms. In vivo studies demonstrated that Tel@PGE facilitated the recruitment of effector CD8+ T lymphocytes (T cells) and the polarization of myeloid-derived suppressor cells (MDSCs) and immunosuppressive M2-like macrophages to tumoricidal antigen-presenting cells. The reshaping of the tumor microenvironment (TME) by Tel@PGE elicited systematic immune responses to significantly prevent B16F10 or 4T-1 tumor postoperative recurrence and metastasis. Therefore, this platform of Tel is expected to provide a clinically available option for effective postoperative combined therapy. STATEMENT OF SIGNIFICANCE: A series of prodrugs or conjugates containing hydrophobic blocks were designed to achieve sustained release at the injection site by reducing the water solubility. However, this strategy sometimes failed short of expectations. Thus, we constructed a biocompatible and biodegradable injectable phase transition gel (PGE) with superior release properties that can be injected subcutaneously into the surgery site. In the long-lasting treatment, the melanoma and breast cancer immunotherapeutic effect significantly enhanced and the risk of cancer metastasis and relapse was reduced. Crucially, for some immune agonists, a superior release control can significantly reduce adverse effects which was decisive for the availability of the drugs.


Subject(s)
Cytokine Release Syndrome , Neoplasm Recurrence, Local , Humans , Adjuvants, Immunologic , Delayed-Action Preparations , Immunotherapy , Toll-Like Receptors/antagonists & inhibitors , Tumor Microenvironment
6.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055015

ABSTRACT

Stereotactic body radiotherapy (SBRT) is known to induce important immunologic changes within the tumor microenvironment (TME). However, little is known regarding the early immune responses within the TME in the first few weeks following SBRT. Therefore, we used the canine spontaneous tumor model to investigate TME responses to SBRT, and how local injection of immune modulatory antibodies to OX40 and TLR 3/9 agonists might modify those responses. Pet dogs with spontaneous cancers (melanoma, carcinoma, sarcoma, n = 6 per group) were randomized to treatment with either SBRT or SBRT combined with local immunotherapy. Serial tumor biopsies and serum samples were analyzed for immunologic responses. SBRT alone resulted at two weeks after treatment in increased tumor densities of CD3+ T cells, FoxP3+ Tregs, and CD204+ macrophages, and increased expression of genes associated with immunosuppression. The addition of OX40/TLR3/9 immunotherapy to SBRT resulted in local depletion of Tregs and tumor macrophages and reduced Treg-associated gene expression (FoxP3), suppressed macrophage-associated gene expression (IL-8), and suppressed exhausted T cell-associated gene expression (CTLA4). Increased concentrations of IL-7, IL-15, and IL-18 were observed in serum of animals treated with SBRT and immunotherapy, compared to animals treated with SBRT. A paradoxical decrease in the density of effector CD3+ T cells was observed in tumor tissues that received combined SBRT and immunotherapy as compared to animals treated with SBRT only. In summary, these results obtained in a spontaneous large animal cancer model indicate that addition of OX40/TLR immunotherapy to SBRT modifies important immunological effects both locally and systemically.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Dog Diseases/therapy , Neoplasms/veterinary , Radiosurgery/methods , Receptors, OX40/antagonists & inhibitors , Toll-Like Receptors/antagonists & inhibitors , Animals , Combined Modality Therapy , Cytokines , Dog Diseases/diagnosis , Dog Diseases/etiology , Dogs , Female , Gene Expression , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Neovascularization, Pathologic/metabolism , Radiotherapy, Image-Guided , Tomography, X-Ray Computed , Treatment Outcome , Tumor Microenvironment/immunology
7.
Adv Sci (Weinh) ; 9(3): e2104051, 2022 01.
Article in English | MEDLINE | ID: mdl-34816630

ABSTRACT

Toll-like receptor (TLR) activation in macrophages plays a critical role in the pathogenesis of acute lung injury (ALI). While TLR inhibition is a promising strategy to control the overwhelming inflammation in ALI, there still lacks effective TLR inhibitors for clinical uses to date. A unique class of peptide-coated gold nanoparticles (GNPs) is previously discovered, which effectively inhibited TLR signaling and protected mice from lipopolysaccharide (LPS)-induced ALI. To fast translate such a discovery into potential clinical applicable nanotherapeutics, herein an elegant strategy of "nano-enabled drug repurposing" with "nano-targeting" is introduced to empower the existing drugs for new uses. Combining transcriptome sequencing with Connectivity Map analysis, it is identified that the proton pump inhibitors (PPIs) share similar mechanisms of action to the discovered GNP-based TLR inhibitor. It is confirmed that PPIs (including omeprazole) do inhibit endosomal TLR signaling and inflammatory responses in macrophages and human peripheral blood mononuclear cells, and exhibits anti-inflammatory activity in an LPS-induced ALI mouse model. The omeprazole is then formulated into a nanoform with liposomes to enhance its macrophage targeting ability and the therapeutic efficacy in vivo. This research provides a new translational strategy of nano-enabled drug repurposing to translate bioactive nanoparticles into clinically used drugs and targeted nano-therapeutics for ALI.


Subject(s)
Acute Lung Injury/drug therapy , Metal Nanoparticles/administration & dosage , Nanomedicine/methods , Proton Pump Inhibitors/pharmacology , Toll-Like Receptors/antagonists & inhibitors , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Proton Pump Inhibitors/metabolism , Toll-Like Receptors/drug effects , Toll-Like Receptors/metabolism
8.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884569

ABSTRACT

Toll-like receptors (TLRs) play a major role in the innate immune system. Several studies have shown the regulatory effects of TLR-mediated pathways on immune and inflammatory diseases. Dysregulated functions of TLRs within the endosomal compartment, including TLR7/9 trafficking, may cause systemic lupus erythematosus (SLE). TLR signaling pathways are fine-tuned by Toll/interleukin-1 receptor (TIR) domain-containing adapters, leading to interferon (IFN)-α production. This study describes a TLR inhibitor peptide 1 (TIP1) that primarily suppresses the downstream signaling mediated by TIR domain-containing adapters in an animal model of lupus and patients with SLE. The expression of most downstream proteins of the TLR7/9/myeloid differentiation factor 88 (MyD88)/IFN regulatory factor 7 signaling was downregulated in major tissues such as the kidney, spleen, and lymph nodes of treated mice. Furthermore, the pathological analysis of the kidney tissue confirmed that TIP1 could improve inflammation in MRL/lpr mice. TIP1 treatment downregulated many downstream proteins associated with TLR signaling, such as MyD88, interleukin-1 receptor-associated kinase, tumor necrosis factor receptor-associated factor 6, and IFN-α, in the peripheral blood mononuclear cells of patients with SLE. In conclusion, our data suggest that TIP1 can serve as a potential candidate for the treatment of SLE.


Subject(s)
Disease Models, Animal , Gene Expression Regulation , Inflammation/prevention & control , Lupus Erythematosus, Systemic/drug therapy , Peptide Fragments/pharmacology , Toll-Like Receptors/antagonists & inhibitors , Animals , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation , Male , Mice , Mice, Inbred MRL lpr , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism
9.
Viruses ; 13(11)2021 10 22.
Article in English | MEDLINE | ID: mdl-34834939

ABSTRACT

The global coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection, threatens the entire world. It has affected every aspect of life and increased the burden on both healthcare and socioeconomic systems. Current studies have revealed that excessive inflammatory immune responses are responsible for the severity of COVID-19, which suggests that anti-inflammatory drugs may be promising therapeutic treatments. However, there are currently a limited number of approved therapeutics for COVID-19. Toll-like receptors (TLRs), which recognize microbial components derived from invading pathogens, are involved in both the initiation of innate responses against SARS-CoV-2 infection and the hyperinflammatory phenotype of COVID-19. In this review, we provide current knowledge on the pivotal role of TLRs in immune responses against SARS-CoV-2 infection and demonstrate the potential effectiveness of TLR-targeting drugs on the control of hyperinflammation in patients with COVID-19.


Subject(s)
COVID-19/immunology , Immunity, Innate , SARS-CoV-2/immunology , Toll-Like Receptors/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , COVID-19/complications , COVID-19/virology , Cytokine Release Syndrome , Humans , SARS-CoV-2/physiology , Severity of Illness Index , Systemic Inflammatory Response Syndrome/complications , Systemic Inflammatory Response Syndrome/drug therapy , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/metabolism , COVID-19 Drug Treatment
10.
Front Immunol ; 12: 704617, 2021.
Article in English | MEDLINE | ID: mdl-34630386

ABSTRACT

Background: The current treatment for HIV-1 is based on blocking various stages in the viral replication cycle using combination antiretroviral therapy (ART). Even though ART effectively controls the infection, it is not curative, and patients must therefore continue treatment life-long. Aim: Here we review recent literature investigating the single or combined effect of toll-like receptor (TLR) agonists and broadly neutralizing antibodies (bNAbs) with the objective to evaluate the evidence for this combination as a means towards an HIV-1 cure. Results: Multiple preclinical studies found significantly enhanced killing of HIV-1 infected cells by TLR agonist-induced innate immune activation or by Fc-mediated effector functions following bNAb administration. However, monotherapy with either agent did not lead to sustained HIV-1 remission in clinical trials among individuals on long-term ART. Notably, findings in non-human primates suggest that a combination of TLR agonists and bNAbs may be able to induce long-term remission after ART cessation and this approach is currently being further investigated in clinical trials. Conclusion: Preclinical findings show beneficial effects of either TLR agonist or bNAb administration for enhancing the elimination of HIV-1 infected cells. Further, TLR agonist-mediated stimulation of innate effector functions in combination with bNAbs may enhance antibody-dependent cellular cytotoxicity and non-human primate studies have shown promising results for this combination strategy. Factors such as immune exhaustion, proviral bNAb sensitivity and time of intervention might impact the clinical success.


Subject(s)
Broadly Neutralizing Antibodies , HIV Infections , HIV-1/immunology , Toll-Like Receptors , Animals , Broadly Neutralizing Antibodies/immunology , Broadly Neutralizing Antibodies/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , Humans , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/immunology
11.
Front Immunol ; 12: 720192, 2021.
Article in English | MEDLINE | ID: mdl-34456928

ABSTRACT

COVID-19 might lead to multi-organ failure and, in some cases, to death. The COVID-19 severity is associated with a "cytokine storm." Danger-associated molecular patterns (DAMPs) are proinflammatory molecules that can activate pattern recognition receptors, such as toll-like receptors (TLRs). DAMPs and TLRs have not received much attention in COVID-19 but can explain some of the gender-, weight- and age-dependent effects. In females and males, TLRs are differentially expressed, likely contributing to higher COVID-19 severity in males. DAMPs and cytokines associated with COVID-19 mortality are elevated in obese and elderly individuals, which might explain the higher risk for severer COVID-19 in these groups. Adenosine signaling inhibits the TLR/NF-κB pathway and, through this, decreases inflammation and DAMPs' effects. As vaccines will not be effective in all susceptible individuals and as new vaccine-resistant SARS-CoV-2 mutants might develop, it remains mandatory to find means to dampen COVID-19 disease severity, especially in high-risk groups. We propose that the regulation of DAMPs via adenosine signaling enhancement might be an effective way to lower the severity of COVID-19 and prevent multiple organ failure in the absence of severe side effects.


Subject(s)
Alarmins/immunology , COVID-19/physiopathology , Inflammation Mediators/immunology , Adenosine/metabolism , Alarmins/antagonists & inhibitors , Animals , COVID-19/complications , COVID-19/immunology , COVID-19/therapy , Humans , Inflammation/prevention & control , Inflammation Mediators/antagonists & inhibitors , Multiple Organ Failure/etiology , Multiple Organ Failure/prevention & control , Patient Acuity , Signal Transduction , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/immunology
12.
J Med Chem ; 64(12): 8010-8041, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34107682

ABSTRACT

Toll-like receptors (TLRs) are members of a large family of evolutionarily conserved pattern recognition receptors (PRRs), which serve as key components of the innate immune system by playing a pivotal role in sensing "nonself" ligands. Endosomal TLRs (TLR3, TLR7, TLR8, and TLR9) can recognize pathogen-derived nucleic acid and initiate an innate immune response because they react against both self- and non-self-origin nucleic acid molecules. Accordingly, both receptor agonists and antagonists are potentially useful in disparate clinical contexts and thus are globally sought after. Recent research has revealed that agonists and antagonists share an overlapping binding region. This Perspective highlights rational medicinal chemistry approaches to elucidate the structural attributes of small molecules capable of agonism or antagonism or of elegantly switching between the two. The structural evolution of different chemotypes can provide the framework for the future development of endosomal TLR agonists and antagonists.


Subject(s)
Heterocyclic Compounds/chemistry , Toll-Like Receptors/agonists , Toll-Like Receptors/antagonists & inhibitors , Animals , Endosomes/chemistry , HEK293 Cells , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Humans , Mice , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Toll-Like Receptors/metabolism
13.
Infect Genet Evol ; 93: 104947, 2021 09.
Article in English | MEDLINE | ID: mdl-34052416

ABSTRACT

Tropical and subtropical areas of the world are affected by leishmaniasis, which is caused by Leishmania spp. It has been categorized as an NTD (neglected tropical disease) because of its negligence. The sand fly of genus Phlebotomus acts as the vector for the transmission of the promastigote form of this protozoan parasite to the mammalian host where it converts to amastigote form in the macrophages. Visceral form of leishmaniasis (VL) is a deadly infection in the endothelial system of the human and other mammals. Only a few chemotherapeutic agents are available for the treatment of this infectious disease whereas no vaccine is available for the control of leishmanial infection. Therefore in the current study, we have tested the effects of gardiquimod (a TLR agonist) as an adjuvant in combination with the formalin-killed antigen of L. donovani as a vaccine. The mice were vaccinated thrice at an interval of 2 weeks and challenged with L. donovani promastigotes after 2 weeks of the last vaccination. We assessed the parasite load, delayed-type hypersensitivity (DTH) responses, humoral and cell-mediated immune response in BALB/c mice before and after challenge infection with L. donovani. Immunized mice were found to have the least parasite load, high DTH response, elevated levels of Th1 cytokines, IgG2a, and nitric oxide than non-immunized and infected control mice. The efficacy of the vaccine was boosted with the use of adjuvant gardiquimod that depicts its potential as an adjuvant in this study. Our study is reporting the adjuvant effects of gardiquimod for the first time. Further studies using other Leishmania species can be performed to signify its role.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Imidazoles/pharmacology , Leishmania donovani/immunology , Leishmaniasis Vaccines/immunology , Leishmaniasis, Visceral/prevention & control , Toll-Like Receptors/antagonists & inhibitors , Animals , Female , Leishmaniasis, Visceral/parasitology , Male , Mice , Mice, Inbred BALB C
14.
Front Immunol ; 12: 653092, 2021.
Article in English | MEDLINE | ID: mdl-33815413

ABSTRACT

Q fever is caused by the obligate intracellular bacterium, Coxiella burnetii, a designated potential agent of bioterrorism because of its route of transmission, resistance to disinfectants, and low infectious dose. The only vaccine licensed for human use is Q-VAX® (Seqirus, licensed in Australia), a formalin-inactivated whole-cell vaccine, which produces severe local and systemic reactogenic responses in previously sensitized individuals. Accordingly, the U.S. Food and Drug Administration and other regulatory bodies around the world, have been reluctant to approve Q-VAX for widespread use. To obviate these adverse reactions, we prepared recombinant protein subunit vaccine candidates containing purified CBU1910, CBU0307, CBU0545, CBU0612, CBU0891, and CBU1398 proteins and TLR triagonist adjuvants. TLR triagonist adjuvants combine different TLR agonists to enhance immune responses to vaccine antigens. We tested both the protective efficacy and reactogenicity of our vaccine candidates in Hartley guinea pigs using intratracheal infection with live C. burnetii. While all of our candidates showed varying degrees of protection during challenge, local reactogenic responses were significantly reduced for one of our vaccine candidates when compared with a formalin-inactivated whole-cell vaccine. Our findings show that subunit vaccines combined with novel TLR triagonist adjuvants can generate protective immunity to C. burnetii infection while reducing reactogenic responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/pharmacology , Coxiella burnetii/immunology , Q Fever/prevention & control , Toll-Like Receptors/antagonists & inhibitors , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/pharmacology , Antigens, Bacterial/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/therapeutic use , Disease Models, Animal , Guinea Pigs , Humans , Immunogenicity, Vaccine , Q Fever/immunology , Q Fever/microbiology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Vaccines, Subunit/genetics , Vaccines, Subunit/pharmacology , Vaccines, Subunit/therapeutic use , Vaccines, Synthetic/genetics , Vaccines, Synthetic/pharmacology , Vaccines, Synthetic/therapeutic use
15.
IUBMB Life ; 73(1): 10-25, 2021 01.
Article in English | MEDLINE | ID: mdl-33217774

ABSTRACT

At the forefront of the battle against pathogens or any endogenously released molecules, toll-like receptors (TLRs) play an important role as the most noble pattern recognition receptors. The ability of these receptors in distinguishing "self" and "non-self" antigens is a cornerstone in the innate immunity system; however, misregulation links inflammatory responses to the development of human cancers. It has been known for some time that aberrant expression and regulation of TLRs not only endows cancer cells an opportunity to escape from the immune system but also supports them through enhancing proliferation and angiogenesis. Over the past decades, cancer research studies have witnessed a number of preclinical and clinical breakthroughs in the field of TLR modulators and some of the agents have exceptionally performed well in advanced clinical trials. In the present review, we have provided a comprehensive review of different TLR agonists and antagonists and discuss their limitations, toxicities, and challenges to outline their future incorporation in cancer treatment strategies.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunity, Innate , Immunotherapy/methods , Neoplasms/drug therapy , Toll-Like Receptors/agonists , Toll-Like Receptors/antagonists & inhibitors , Animals , Humans , Neoplasms/immunology , Neoplasms/pathology , Signal Transduction
16.
HNO ; 68(12): 916-921, 2020 Dec.
Article in German | MEDLINE | ID: mdl-33128107

ABSTRACT

BACKGROUND: CD8+ cells are key players in the identification and elimination of cancer cells. Cancers can escape an effective T cell response by inducing an exhausted cell state, which limits the cytotoxic capacity of the effector cells. Among other mechanisms, new checkpoint inhibitors reactivate exhausted, dysfunctional T cells. CD8+ T cells can eliminate tumor cells after presentation of tumor-specific antigens via antigen-presenting cells (APCs). APC-mediated tumor recognition is mainly stimulated by Toll-like receptors (TLRs). OBJECTIVE: This study investigates the effect of TLR agonists on APCs as well as stimulatory and inhibitory signaling pathways of the T cell-APC interaction. MATERIALS AND METHODS: Gene expression of interleukin (IL)12 and programmed death ligand 1 (PD-L1) was analyzed by quantitative polymerase chain reaction (qPCR) after 0, 8, 24, and 48 h of CD14+ cell stimulation with CpG. Protein expression of inhibitor of nuclear factor kappa B (IκBα) after CpG stimulation was investigated by western blot. CD8+ T cells were stimulated for 72 h with or without programmed cell death protein 1 (PD-1) checkpoint blockade and analyzed for expression of PD­1, Tim­3, CTLA4, and Lag3 by flow cytometry. RESULTS: TLR stimulation (by unmethylated CpG DNA) of APCs upregulates immunostimulatory signals such as IL12 expression but also activates immunoinhibitory signaling pathways such as PD-L1 expression. This signaling is NF-κB dependent. After blockade of the PD-1/PD-L1 signaling pathway, overexpression of other immune checkpoint inhibitory receptors was observed-a potential explanation for lacking therapeutic responses after TLR stimulation with PD­1 checkpoint blockade. CONCLUSION: TLR stimulation causes APCs in the tumor microenvironment to upregulate PD-L1 in an NF-κB-mediated fashion, thereby contributing to CD8+ T cell exhaustion. The effect of PD­1 blockade after TLR stimulation might be impaired due to upregulation of other checkpoint inhibitors.


Subject(s)
Antigen-Presenting Cells , CD8-Positive T-Lymphocytes , Signal Transduction , Toll-Like Receptors , B7-H1 Antigen/metabolism , NF-kappa B/physiology , Toll-Like Receptors/antagonists & inhibitors , Tumor Microenvironment
17.
J Med Chem ; 63(22): 13466-13513, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32845153

ABSTRACT

Toll-like receptors (TLRs) are a class of proteins that recognize pathogen-associated molecular patterns (PAMPs) and damaged-associated molecular patterns (DAMPs), and they are involved in the regulation of innate immune system. These transmembrane receptors, localized at the cellular or endosomal membrane, trigger inflammatory processes through either myeloid differentiation primary response 88 (MyD88) or TIR-domain-containing adapter-inducing interferon-ß (TRIF) signaling pathways. In the last decades, extensive research has been performed on TLR modulators and their therapeutic implication under several pathological conditions, spanning from infections to cancer, from metabolic disorders to neurodegeneration and autoimmune diseases. This Perspective will highlight the recent discoveries in this field, emphasizing the role of TLRs in different diseases and the therapeutic effect of their natural and synthetic modulators, and it will discuss insights for the future exploitation of TLR modulators in human health.


Subject(s)
Drug Delivery Systems/trends , Immunity, Innate/physiology , Toll-Like Receptors/agonists , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/immunology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Communicable Diseases/drug therapy , Communicable Diseases/immunology , Communicable Diseases/metabolism , Drug Delivery Systems/methods , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemistry , Immunity, Innate/drug effects , Metabolic Diseases/drug therapy , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Protein Structure, Secondary , Toll-Like Receptors/metabolism
18.
Cells ; 9(7)2020 07 09.
Article in English | MEDLINE | ID: mdl-32660060

ABSTRACT

Toll-like receptors (TLRs) play a fundamental role in the inflammatory response against invading pathogens. However, the dysregulation of TLR-signaling pathways is implicated in several autoimmune/inflammatory diseases. Here, we show that a novel small molecule TLR-inhibitor (TAC5) and its derivatives TAC5-a, TAC5-c, TAC5-d, and TAC5-e predominantly antagonized poly(I:C) (TLR3)-, imiquimod (TLR7)-, TL8-506 (TLR8)-, and CpG-oligodeoxynucleotide (TLR9)-induced signaling pathways. TAC5 and TAC5-a significantly hindered the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), reduced the phosphorylation of mitogen-activated protein kinases, and inhibited the secretion of tumor necrosis factor-α (TNF-α) and interleukin-6. Besides, TAC5-a prevented the progression of psoriasis and systemic lupus erythematosus (SLE) in mice. Interestingly, TAC5 and TAC5-a did not affect Pam3CSK4 (TLR1/2)-, FSL-1 (TLR2/6)-, or lipopolysaccharide (TLR4)-induced TNF-α secretion, indicating their specificity towards endosomal TLRs (TLR3/7/8/9). Collectively, our data suggest that the TAC5 series of compounds are potential candidates for treating autoimmune diseases such as psoriasis or SLE.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Immunologic Factors/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Psoriasis/drug therapy , Toll-Like Receptors/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Binding Sites , Endosomes/metabolism , Female , Immunologic Factors/chemistry , Immunologic Factors/therapeutic use , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , NF-kappa B/metabolism , Protein Binding , Quantitative Structure-Activity Relationship , RAW 264.7 Cells , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Toll-Like Receptors/chemistry , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Front Immunol ; 11: 1236, 2020.
Article in English | MEDLINE | ID: mdl-32625214

ABSTRACT

Toll like receptors (TLRs) are a family of pattern recognition receptors that play a central role in the innate immune response. These receptors are expressed on a wide variety of immune and non-immune cells, and they help shape the immune response to infection and injury through the recognition of pathogen-associated molecular patterns (PAMPs) as well as endogenous damage-associated molecular patterns (DAMPs). Accumulating evidence suggests that, in addition to regulating mature effector immune cells, TLRs can influence the immune response from the level of the hematopoietic stem cell (HSC). HSCs express TLRs, and exposure to TLR ligands influences the cycling, differentiation, and function of HSCs, with chronic TLR stimulation leading to impairment of normal HSC repopulating activity. Moreover, enhanced TLR expression and signaling is associated with myelodysplastic syndromes (MDS), a heterogenous group of HSC disorders characterized by ineffective hematopoiesis and a high risk of transformation to acute leukemias. In this review, we will discuss the role of TLR signaling in the pathogenesis of MDS, focusing on the known direct and indirect effects of this type of signaling on HSCs, the mechanisms of TLR signaling upregulation in MDS, the changes in TLR expression with disease progression, and the therapeutic implications for modulating TLR signaling in the treatment of MDS.


Subject(s)
Disease Susceptibility , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Animals , Cell Death , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Humans , Molecular Targeted Therapy , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/drug therapy , Severity of Illness Index , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/genetics
20.
Curr Atheroscler Rep ; 22(5): 19, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32458165

ABSTRACT

PURPOSE OF REVIEW: In this review, we discuss the evidence supporting the effects of statins on mast cells (MCs) in atherosclerosis and their molecular mechanism of action. RECENT FINDINGS: Statins or HMG-CoA reductase inhibitors are known for their lipid-lowering properties and are widely used in the prevention and treatment of cardiovascular diseases. There is growing evidence that statins have an inhibitory effect on MCs, which contributes to the pleiotropic effect of statins in various diseases. MCs are one of the crucial effectors of the immune system which play an essential role in the pathogenesis of multiple disorders. Recent studies have shown that MCs are involved in the development of atherosclerotic plaques. MCs secrete various inflammatory cytokines (IL-6, IL4, TNF-α, and IFNγ) and inflammatory mediators (histamine, tryptase, proteoglycans) after activation by various stimulants. This, in turn, will exacerbate atherosclerosis. Statins suppress the activation of MCs via IgE inhibition which leads to inhibition of inflammatory mediators and cytokines which are involved in the development and progression of atherosclerosis. In keeping with this evidence presented here, MCs can be considered as one of the therapeutic targets for statins in the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/physiopathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mast Cells/drug effects , Animals , Cell Degranulation/drug effects , Cytokines/metabolism , Histamine Release/drug effects , Humans , Mast Cells/immunology , Mast Cells/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors/therapeutic use , Phospholipase A2 Inhibitors/pharmacology , Phospholipase A2 Inhibitors/therapeutic use , Plaque, Atherosclerotic/physiopathology , Receptors, LDL/agonists , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL