Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
1.
Front Immunol ; 14: 1050113, 2023.
Article in English | MEDLINE | ID: mdl-36865548

ABSTRACT

Introduction: Sulfavant A (SULF A) is a synthetic derivative of naturally occurring sulfolipids. The molecule triggers TREM2-related maturation of dendritic cells (DCs) and has shown promising adjuvant activity in a cancer vaccine model. Methods: the immunomodulatory activity of SULF A is tested in an allogeneic mixed lymphocyte reaction (MLR) assay based on monocyte-derived dendritic cells and naïve T lymphocytes from human donors. Flow cytometry multiparametric analyses and ELISA assays were performed to characterize the immune populations, T cell proliferation, and to quantify key cytokines. Results: Supplementation of 10 µg/mL SULF A to the co-cultures induced DCs to expose the costimulatory molecules ICOSL and OX40L and to reduce release of the pro-inflammatory cytokine IL-12. After 7 days of SULF A treatment, T lymphocytes proliferated more and showed increased IL-4 synthesis along with downregulation of Th1 signals such as IFNγ, T-bet and CXCR3. Consistent with these findings, naïve T cells polarized toward a regulatory phenotype with up-regulation of FOXP3 expression and IL-10 synthesis. Flow cytometry analysis also supported the priming of a CD127-/CD4+/CD25+ subpopulation positive for ICOS, the inhibitory molecule CTLA-4, and the activation marker CD69. Discussion: These results prove that SULF A can modulate DC-T cell synapse and stimulate lymphocyte proliferation and activation. In the hyperresponsive and uncontrolled context of the allogeneic MLR, the effect is associated to differentiation of regulatory T cell subsets and dampening of inflammatory signals.


Subject(s)
Adjuvants, Immunologic , Hematopoietic Stem Cell Transplantation , Membrane Glycoproteins , Receptors, Immunologic , Humans , Adjuvants, Immunologic/pharmacology , Interleukin-12 , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/agonists , Receptors, Immunologic/agonists
2.
J Neuroinflammation ; 19(1): 289, 2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36463233

ABSTRACT

BACKGROUND: Neuroinflammation is one of the most important processes in secondary injury after traumatic brain injury (TBI). Triggering receptor expressed on myeloid cells 2 (TREM2) has been proven to exert neuroprotective effects in neurodegenerative diseases and stroke by modulating neuroinflammation, and promoting phagocytosis and cell survival. However, the role of TREM2 in TBI has not yet been elucidated. In this study, we are the first to use COG1410, an agonist of TREM2, to assess the effects of TREM2 activation in a murine TBI model. METHODS: Adult male wild-type (WT) C57BL/6 mice and adult male TREM2 KO mice were subjected to different treatments. TBI was established by the controlled cortical impact (CCI) method. COG1410 was delivered 1 h after CCI via tail vein injection. Western blot analysis, immunofluorescence, laser speckle contrast imaging (LSCI), neurological behaviour tests, brain electrophysiological monitoring, Evans blue assays, magnetic resonance imaging (MRI), and brain water content measurement were performed in this study. RESULTS: The expression of endogenous TREM2 peaked at 3 d after CCI, and it was mainly expressed on microglia and neurons. We found that COG1410 improved neurological functions within 3 d, as well as neurological functions and brain electrophysiological activity at 2 weeks after CCI. COG1410 exerted neuroprotective effects by inhibiting neutrophil infiltration and microglial activation, and suppressing neuroinflammation after CCI. In addition, COG1410 treatment alleviated blood brain barrier (BBB) disruption and brain oedema; furthermore, COG1410 promoted cerebral blood flow (CBF) recovery at traumatic injury sites after CCI. In addition, COG1410 suppressed neural apoptosis at 3 d after CCI. TREM2 activation upregulated p-Akt, p-CREB, BDNF, and Bcl-2 and suppressed TNF-α, IL-1ß, Bax, and cleaved caspase-3 at 3 d after CCI. Moreover, TREM2 knockout abolished the effects of COG1410 on vascular phenotypes and microglial states. Finally, the neuroprotective effects of COG1410 were suppressed by TREM2 depletion. CONCLUSIONS: Altogether, we are the first to demonstrate that TREM2 activation by COG1410 alleviated neural damage through activation of Akt/CREB/BDNF signalling axis in microglia after CCI. Finally, COG1410 treatment improved neurological behaviour and brain electrophysiological activity after CCI.


Subject(s)
Brain Injuries, Traumatic , Animals , Male , Mice , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/immunology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/immunology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Immunologic/agonists , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Disease Models, Animal , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/immunology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Nervous System/drug effects , Nervous System/immunology
3.
J Control Release ; 347: 476-488, 2022 07.
Article in English | MEDLINE | ID: mdl-35577151

ABSTRACT

Despite success in vaccinating populations against SARS-CoV-2, concerns about immunity duration, continued efficacy against emerging variants, protection from infection and transmission, and worldwide vaccine availability remain. Molecular adjuvants targeting pattern recognition receptors (PRRs) on antigen-presenting cells (APCs) could improve and broaden the efficacy and durability of vaccine responses. Native SARS-CoV-2 infection stimulates various PRRs, including toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors. We hypothesized that targeting PRRs using molecular adjuvants on nanoparticles (NPs) along with a stabilized spike protein antigen could stimulate broad and efficient immune responses. Adjuvants targeting TLR4 (MPLA), TLR7/8 (R848), TLR9 (CpG), and RIG-I (PUUC) delivered on degradable polymer NPs were combined with the S1 subunit of spike protein and assessed in vitro with isogeneic mixed lymphocyte reactions (isoMLRs). For in vivo studies, the adjuvant-NPs were combined with stabilized spike protein or spike-conjugated NPs and assessed using a two-dose intranasal or intramuscular vaccination model in mice. Combination adjuvant-NPs simultaneously targeting TLR and RIG-I receptors (MPLA+PUUC, CpG+PUUC, and R848+PUUC) differentially induced T cell proliferation and increased proinflammatory cytokine secretion by APCs in vitro. When delivered intranasally, MPLA+PUUC NPs enhanced CD4+CD44+ activated memory T cell responses against spike protein in the lungs while MPLA NPs increased anti-spike IgA in the bronchoalveolar (BAL) fluid and IgG in the blood. Following intramuscular delivery, PUUC NPs induced strong humoral immune responses, characterized by increases in anti-spike IgG in the blood and germinal center B cell populations (GL7+ and BCL6+ B cells) in the draining lymph nodes (dLNs). MPLA+PUUC NPs further boosted spike protein-neutralizing antibody titers and T follicular helper cell populations in the dLNs. These results suggest that protein subunit vaccines with particle-delivered molecular adjuvants targeting TLR4 and RIG-I could lead to robust and unique route-specific adaptive immune responses against SARS-CoV-2.


Subject(s)
COVID-19 Vaccines , COVID-19 , DEAD Box Protein 58 , Nanoparticles , Receptors, Immunologic , Toll-Like Receptor 4 , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Drug Delivery Systems , Immunity, Humoral , Immunoglobulin G , Mice , Nanoparticles/chemistry , Receptors, Immunologic/agonists , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Toll-Like Receptor 4/agonists
4.
Front Immunol ; 12: 729189, 2021.
Article in English | MEDLINE | ID: mdl-34603303

ABSTRACT

Several SARS-CoV-2 vaccines have received EUAs, but many issues remain unresolved, including duration of conferred immunity and breadth of cross-protection. Adjuvants that enhance and shape adaptive immune responses that confer broad protection against SARS-CoV-2 variants will be pivotal for long-term protection as drift variants continue to emerge. We developed an intranasal, rationally designed adjuvant integrating a nanoemulsion (NE) that activates TLRs and NLRP3 with an RNA agonist of RIG-I (IVT DI). The combination adjuvant with spike protein antigen elicited robust responses to SARS-CoV-2 in mice, with markedly enhanced TH1-biased cellular responses and high virus-neutralizing antibody titers towards both homologous SARS-CoV-2 and a variant harboring the N501Y mutation shared by B1.1.7, B.1.351 and P.1 variants. Furthermore, passive transfer of vaccination-induced antibodies protected naive mice against heterologous viral challenge. NE/IVT DI enables mucosal vaccination, and has the potential to improve the immune profile of a variety of SARS-CoV-2 vaccine candidates to provide effective cross-protection against future drift variants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Viral/immunology , COVID-19 Vaccines/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , Adaptive Immunity/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Chlorocebus aethiops , Cross Protection/immunology , DEAD Box Protein 58 , HEK293 Cells , Humans , Immunity, Humoral/immunology , Immunization, Passive , Mice , Mice, Inbred C57BL , Receptors, Immunologic/agonists , Recombinant Proteins/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Vero Cells
5.
Respir Res ; 22(1): 262, 2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34620168

ABSTRACT

BACKGROUND: Prostaglandin D2 (PGD2) signaling via prostaglandin D2 receptor 2 (DP2) contributes to atopic and non-atopic asthma. Inhibiting DP2 has shown therapeutic benefit in certain subsets of asthma patients, improving eosinophilic airway inflammation. PGD2 metabolites prolong the inflammatory response in asthmatic patients via DP2 signaling. The role of PGD2 metabolites on eosinophil and ILC2 activity is not fully understood. METHODS: Eosinophils and ILC2s were isolated from peripheral blood of atopic asthmatic patients. Eosinophil shape change, ILC2 migration and IL-5/IL-13 cytokine secretion were measured after stimulation with seven PGD2 metabolites in presence or absence of the selective DP2 antagonist fevipiprant. RESULTS: Selected metabolites induced eosinophil shape change with similar nanomolar potencies except for 9α,11ß-PGF2. Maximal values in forward scatter of eosinophils were comparable between metabolites. ILC2s migrated dose-dependently in the presence of selected metabolites except for 9α,11ß-PGF2 with EC50 values ranging from 17.4 to 91.7 nM. Compared to PGD2, the absolute cell migration was enhanced in the presence of Δ12-PGD2, 15-deoxy-Δ12,14-PGD2, PGJ2, Δ12-PGJ2 and 15-deoxy-Δ12,14-PGJ2. ILC2 cytokine production was dose dependent as well but with an average sixfold reduced potency compared to cell migration (IL-5 range 108.1 to 526.9 nM, IL-13 range: 125.2 to 788.3 nM). Compared to PGD2, the absolute cytokine secretion was reduced in the presence of most metabolites. Fevipiprant dose-dependently inhibited eosinophil shape change, ILC2 migration and ILC2 cytokine secretion with (sub)-nanomolar potencies. CONCLUSION: Prostaglandin D2 metabolites initiate ILC2 migration and IL-5 and IL-13 cytokine secretion in a DP2 dependent manner. Our data indicate that metabolites may be important for in vivo eosinophil activation and ILC2 migration and to a lesser extent for ILC2 cytokine secretion.


Subject(s)
Asthma/drug therapy , Eosinophils/drug effects , Lymphocytes/drug effects , Prostaglandin D2/pharmacology , Receptors, Immunologic/agonists , Receptors, Prostaglandin/agonists , Adolescent , Adult , Aged , Asthma/immunology , Asthma/metabolism , Cell Movement/drug effects , Cell Shape/drug effects , Cells, Cultured , Eosinophils/immunology , Eosinophils/metabolism , Female , Humans , Indoleacetic Acids/pharmacology , Interleukin-13/metabolism , Interleukin-5/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Middle Aged , Prostaglandin Antagonists/pharmacology , Prostaglandin D2/analogs & derivatives , Pyridines/pharmacology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Signal Transduction , Young Adult
6.
Exp Mol Med ; 53(2): 281-290, 2021 02.
Article in English | MEDLINE | ID: mdl-33594256

ABSTRACT

Synucleinopathies are age-related neurological disorders characterized by the progressive deposition of α-synuclein (α-syn) aggregates and include Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Although cell-to-cell α-syn transmission is thought to play a key role in the spread of α-syn pathology, the detailed mechanism is still unknown. Neuroinflammation is another key pathological feature of synucleinopathies. Previous studies have identified several immune receptors that mediate neuroinflammation in synucleinopathies, such as Toll-like receptor 2 (TLR2). However, the species of α-syn aggregates varies from study to study, and how different α-syn aggregate species interact with innate immune receptors has yet to be addressed. Therefore, we investigated whether innate immune receptors can facilitate the uptake of different species of α-syn aggregates. Here, we examined whether stimulation of TLRs could modulate the cellular uptake and degradation of α-syn fibrils despite a lack of direct interaction. We observed that stimulation of TLR2 in vitro accelerated α-syn fibril uptake in neurons and glia while delaying the degradation of α-syn in neurons and astrocytes. Internalized α-syn was rapidly degraded in microglia regardless of whether TLR2 was stimulated. However, cellular α-syn uptake and degradation kinetics were not altered by TLR4 stimulation. In addition, upregulation of TLR2 expression in a synucleinopathy mouse model increased the density of Lewy-body-like inclusions and induced morphological changes in microglia. Together, these results suggest that cell type-specific modulation of TLR2 may be a multifaceted and promising therapeutic strategy for synucleinopathies; inhibition of neuronal and astroglial TLR2 decreases pathogenic α-syn transmission, but activation of microglial TLR2 enhances microglial extracellular α-syn clearance.


Subject(s)
Brain/immunology , Brain/metabolism , Immunity, Innate , Receptors, Immunologic/agonists , Receptors, Immunologic/metabolism , alpha-Synuclein/metabolism , Animals , Astrocytes , Brain/pathology , Cell Line , Extracellular Space/metabolism , Humans , Immunity, Innate/genetics , Immunohistochemistry , Mice , Mice, Transgenic , Microglia/metabolism , Models, Animal , Models, Biological , Neurons/metabolism , Protease Inhibitors/pharmacology , Protein Binding , Protein Transport , Proteolysis , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , alpha-Synuclein/genetics
7.
ChemMedChem ; 16(8): 1246-1251, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33415819

ABSTRACT

Mincle agonists have been shown to induce inflammatory cytokine production, such as tumor necrosis factor-alpha (TNF) and promote the development of a Th1/Th17 immune response that might be crucial to development of effective vaccination against pathogens such as Mycobacterium tuberculosis. As an expansion of our previous work, a library of 6,6'-amide and sulfonamide α,α-d-trehalose compounds with various substituents on the aromatic ring was synthesized efficiently in good to excellent yields. These compounds were evaluated for their ability to activate the human C-type lectin receptor Mincle by the induction of cytokines from human peripheral blood mononuclear cells. A preliminary structure-activity relationship (SAR) of these novel trehalose diamides and sulfonamides revealed that aryl amide-linked trehalose compounds demonstrated improved activity and relatively high potency cytokine production compared to the Mincle ligand trehalose dibehenate adjuvant (TDB) and the natural ligand trehalose dimycolate (TDM) inducing dose-dependent and human-Mincle-specific stimulation in a HEK reporter cell line.


Subject(s)
Adjuvants, Immunologic/pharmacology , Lectins, C-Type/agonists , Receptors, Immunologic/agonists , Sulfonamides/pharmacology , Trehalose/analogs & derivatives , Trehalose/pharmacology , Adjuvants, Immunologic/chemical synthesis , HEK293 Cells , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Sulfonamides/chemical synthesis , Tumor Necrosis Factor-alpha/metabolism
8.
Int J Mol Sci ; 22(1)2021 Jan 03.
Article in English | MEDLINE | ID: mdl-33401559

ABSTRACT

Retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) mediate anti-viral response through mitochondria. In addition, RLR activation induces anti-tumor effects on various cancers. We previously reported that the RLR agonist Poly(I:C)-HMW/LyoVec™ (Poly(I:C)) enhanced radiosensitivity and that cotreatment with Poly(I:C) and ionizing radiation (IR) more than additively increased cell death in lung adenocarcinoma cells, indicating that Poly(I:C) modulates the cellular radiation response. However, it remains unclear how mitochondria are involved in the modulation of this response. Here, we investigated the involvement of mitochondrial dynamics and mitochondrial ribosome protein death-associated protein 3 (DAP3) in the modulation of cellular radiation response by Poly(I:C) in A549 and H1299 human lung adenocarcinoma cell lines. Western blotting revealed that Poly(I:C) decreased the expression of mitochondrial dynamics-related proteins and DAP3. In addition, siRNA experiments showed that DAP3, and not mitochondrial dynamics, is involved in the resistance of lung adenocarcinoma cells to IR-induced cell death. Finally, we revealed that a more-than-additive effect of cotreatment with Poly(I:C) and IR on increasing cell death was diluted by DAP3-knockdown because of an increase in cell death induced by IR alone. Together, our findings suggest that RLR agonist Poly(I:C) modulates the cellular radiation response of lung adenocarcinoma cells by downregulating DAP3 expression.


Subject(s)
Adenocarcinoma of Lung/pathology , Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Poly I-C/pharmacology , RNA-Binding Proteins/metabolism , Radiation, Ionizing , Receptors, Immunologic/agonists , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/radiotherapy , Apoptosis , Apoptosis Regulatory Proteins/genetics , Cell Proliferation , DEAD Box Protein 58 , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , RNA-Binding Proteins/genetics , Tumor Cells, Cultured
9.
Int Immunopharmacol ; 91: 107215, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33348294

ABSTRACT

The survivability of Mycobacterium tuberculosis (M.tb) in macrophages in granuloma is a predominant cause for tuberculosis (TB) infection and recurrence. However, the mechanism of mycobacterial clearance in macrophages still needs further study. Here, we explored a novel role of B and T lymphocyte Attenuator (BTLA) in macrophage-mediated host defense against mycobacterial infection. We found that the surface expression of BTLA was increased in CD14+ monocytes from active TB patients. The mRNA levels of BTLA were induced in human and mice monocytes/macrophages during Mycobacterium bovis BCG or M.tb H37Rv infection, as well as spleen and lung of H37Rv-infected mice. Furthermore, silencing of BTLA promoted the intracellular survival of BCG and H37Rv by suppressing the autophagy in macrophages but not effecting phagocytosis, reactive oxygen species (ROS) and apoptosis. Silence of BTLA reduced bacterial-autophagosome and bacterial-lysosome colocalization. Moreover, BTLA inhibited AKT and mTOR signaling substrates S6K and 4EBP1 phosphorylation in BCG and H37Rv infected macrophages, and BTLA-mediated AKT-mTOR signaling and intracellular BCG survival were reversed by PI3K inhibitors in macrophages. Finally, treatment with BTLA agonist ameliorated lung pathology and promoted autophagy and mycobacterial clearance during mycobacterial infection in vivo. These results demonstrate that BTLA promotes host defense against mycobacteria by enhancing autophagy, which may provide potential therapeutic interventions against tuberculosis.


Subject(s)
Autophagy , Lung/enzymology , Macrophages/enzymology , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Immunologic/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberculosis, Pulmonary/enzymology , Animals , Antitubercular Agents/pharmacology , Autophagy/drug effects , Disease Models, Animal , Host-Pathogen Interactions , Humans , Lung/drug effects , Lung/immunology , Lung/microbiology , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Nude , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , RAW 264.7 Cells , Receptors, Immunologic/agonists , Receptors, Immunologic/genetics , Signal Transduction , THP-1 Cells , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/prevention & control
10.
JCI Insight ; 5(18)2020 09 01.
Article in English | MEDLINE | ID: mdl-32870822

ABSTRACT

Despite advances in identifying the key immunoregulatory roles of many of the human leukocyte immunoglobulin-like receptor (LILR) family members, the function of the inhibitory molecule LILRB3 (ILT5, CD85a, LIR3) remains unclear. Studies indicate a predominant myeloid expression; however, high homology within the LILR family and a relative paucity of reagents have hindered progress toward identifying the function of this receptor. To investigate its function and potential immunomodulatory capacity, a panel of LILRB3-specific monoclonal antibodies (mAbs) was generated. LILRB3-specific mAbs bound to discrete epitopes in Ig-like domain 2 or 4. LILRB3 ligation on primary human monocytes by an agonistic mAb resulted in phenotypic and functional changes, leading to potent inhibition of immune responses in vitro, including significant reduction in T cell proliferation. Importantly, agonizing LILRB3 in humanized mice induced tolerance and permitted efficient engraftment of allogeneic cells. Our findings reveal powerful immunosuppressive functions of LILRB3 and identify it as an important myeloid checkpoint receptor.


Subject(s)
Antigens, CD/genetics , Epitopes/immunology , Immune Checkpoint Proteins/genetics , Immune Tolerance , Lymphoma/genetics , Monocytes/immunology , Receptors, Immunologic/genetics , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , Antigens, CD/immunology , Cell Line, Tumor , Cell Proliferation , Epitope Mapping , Epitopes/chemistry , Gene Expression Profiling , Gene Expression Regulation , Heterografts , Humans , Immune Checkpoint Proteins/immunology , Lymphoma/immunology , Lymphoma/mortality , Lymphoma/pathology , Mice , Monocytes/cytology , Peptide Library , Primary Cell Culture , Receptors, Immunologic/agonists , Receptors, Immunologic/immunology , Survival Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transplantation, Homologous
11.
Curr Opin Immunol ; 67: 1-9, 2020 12.
Article in English | MEDLINE | ID: mdl-32619929

ABSTRACT

Central and peripheral tolerance both contribute to protection against autoimmunity. The pathogenesis of autoimmunity, however, can result from critical deficits or limitations in peripheral and/or central tolerance mechanisms, presenting an opportunity for therapeutic intervention. Recent advances highlight the substantial impact of inhibitory receptors (IRs), which mediate peripheral tolerance, in autoimmunity. Deletion and blockade studies in mice, IR disruption in humans, and correlation with positive disease outcomes all highlight potential clinical benefits of enhancing IR signaling (agonism)-specifically CTLA4, PD1, LAG3, TIM3 and TIGIT-to treat autoimmune disease. Although critical questions remain, IR agonists represent an unappreciated and untapped opportunity for the treatment of autoimmune and inflammatory diseases.


Subject(s)
Autoimmune Diseases/drug therapy , Receptors, Immunologic/agonists , Animals , Autoimmune Diseases/immunology , Humans , Signal Transduction/immunology
12.
Org Biomol Chem ; 18(19): 3659-3663, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32356529

ABSTRACT

Ac1PIM1 is a potential biosynthetic intermediate for phosphatidylinositol mannosides (PIMs) from Mycobacterium tuberculosis. We achieved the first synthesis of Ac1PIM1 by utilizing an allyl-type protecting group strategy and regioselective phosphorylation of inositol. A very potent agonist of an innate immune receptor DCAR, which is better than previously known agonists, is demonstrated.


Subject(s)
Immunomodulation/drug effects , Lectins, C-Type/agonists , Mycobacterium tuberculosis/chemistry , Phosphatidylinositols/pharmacology , Receptors, Immunologic/agonists , Animals , Cytokines/biosynthesis , Lectins, C-Type/immunology , Mice , Mycobacterium tuberculosis/immunology , Phosphatidylinositols/chemical synthesis , Phosphatidylinositols/chemistry , Phosphorylation , RAW 264.7 Cells , Receptors, Immunologic/immunology
13.
ACS Chem Biol ; 15(7): 1835-1841, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32293864

ABSTRACT

The first asymmetric total synthesis of three structures proposed for mycobacterial diacyl trehaloses, DAT1, DAT2, and DAT3 is reported. The presence of two of these glycolipids, DAT1 and DAT3, within different strains of pathogenic M. tuberculosis was confirmed, and it was shown that their abundance varies significantly. In mass spectrometry, synthetic DAT2 possessed almost identical fragmentation patterns to presumptive DAT2 from Mycobacterium tuberculosis H37Rv, but did not coelute by HPLC, raising questions as the precise relationship of the synthetic and natural materials. The synthetic DATs were examined as agonists for signaling by the C-type lectin, Mincle. The small differences in the chemical structure of the lipidic parts of DAT1, DAT2, and DAT3 led to drastic differences of Mincle binding and activation, with DAT3 showing similar potency as the known Mincle agonist trehalose dimycolate (TDM). In the future, DAT3 could serve as basis for the design of vaccine adjuvants with simplified chemical structure.


Subject(s)
Glycolipids/pharmacology , Lectins, C-Type/agonists , Membrane Proteins/agonists , Receptors, Immunologic/agonists , Trehalose/analogs & derivatives , Trehalose/pharmacology , Animals , Chromatography, Liquid , Glycolipids/chemical synthesis , Glycolipids/isolation & purification , Humans , Mass Spectrometry , Mice , Molecular Structure , Mycobacterium tuberculosis/chemistry , Protein Binding , Stereoisomerism , Trehalose/isolation & purification
14.
Chem Commun (Camb) ; 56(31): 4292-4295, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32182321

ABSTRACT

The innate immune receptor Mincle senses lipid-based molecules derived from pathogens, commensals and altered self. Based on emerging structure-activity relationships we design simple alkyl 6-O-acyl-ß-d-glucosides that are effective agonists of Mincle and signal with potency on par with the prototypical ligand trehalose dimycolate.


Subject(s)
Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Glucosides/pharmacology , Lectins, C-Type/agonists , Receptors, Immunologic/agonists , Signal Transduction/drug effects , Animals , Glucosides/chemical synthesis , Humans , Mice
15.
Curr Cancer Drug Targets ; 20(5): 372-381, 2020.
Article in English | MEDLINE | ID: mdl-31951181

ABSTRACT

BACKGROUND: Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) play key roles in the antiviral response, but recent works show that RLR activation elicits anticancer activity as well, including apoptosis. Previously, we demonstrated that the anticancer activity of the RLR agonist Poly(I:C)-HMW/LyoVec™ [Poly(I:C)-HMW] against human lung cancer cells was enhanced by cotreatment with ionizing radiation (IR). In addition, cotreatment with Poly(I:C)-HMW and IR induced apoptosis in a Fas-independent manner, and increased Fas expression on the cell surface. OBJECTIVE: The current study investigated the resultant hypothesis that Fas ligand (FasL) may enhance apoptosis in lung cancer cells cotreated with Poly(I:C)-HMW+IR. METHODS: FasL was added into culture medium at 24 h following cotreatment with Poly(I:C)- HMW+IR, after upregulation of cell surface Fas expression on human lung cancer cells A549 and H1299 have already been discussed. RESULTS: FasL enhanced the apoptosis of A549 and H1299 cells treated with Poly(I:C)-HMW+IR. Similarly, IR alone - and not Poly(I:C)-HMW - resulted in the upregulation of cell surface Fas expression followed by a high response to FasL-induced apoptosis, thus suggesting that the high sensitivity of cells treated with Poly(I:C)-HMW+IR to FasL-induced apoptosis resulted from the cellular response to IR. Finally, knockdown of Fas by siRNA confirmed that the high response of treated cells to FasL-induced apoptosis is dependent on Fas expression. CONCLUSION: In summary, the present study indicates that upregulated Fas expression following cotreatment with Poly(I:C)-HMW and IR is responsive to FasL-induced apoptosis, and a combination of RLR agonist, IR, and FasL could be a potential promising cancer therapy.


Subject(s)
Antiviral Agents/pharmacology , Apoptosis , Chemoradiotherapy/methods , Fas Ligand Protein/metabolism , Lung Neoplasms/pathology , Poly I-C/pharmacology , Receptors, Immunologic/agonists , Cell Proliferation , DEAD Box Protein 58 , Fas Ligand Protein/genetics , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Tumor Cells, Cultured
16.
Pharmacol Res ; 154: 104192, 2020 04.
Article in English | MEDLINE | ID: mdl-30836160

ABSTRACT

Cancer immunotherapy has come of age with the advent of immune checkpoint inhibitors. In this article we review how agonists for receptors of the innate immune system, the Toll-like receptors and the RIG-I-like receptors, impact anticancer immune responses. Treatment with these agonists enhances the activity of anticancer effector cells, such as cytotoxic T cells and NK cells, and at the same time blocks the activity of immunosuppressive cell types such as regulatory T cells and myeloid-derived suppressor cells. These compounds also impact the recruitment of immune cells to the tumor. The phenomena of pattern-recognition receptor tolerance and reprogramming and their implications for immunotherapy are discussed. Finally, novel delivery systems that target the immune-stimulating drugs to the tumor or the tumor-draining lymph nodes to enhance their efficacy and safety are presented.


Subject(s)
Immunotherapy , Neoplasms/therapy , Receptors, Immunologic/agonists , Toll-Like Receptors/agonists , Animals , DEAD Box Protein 58/immunology , Humans , Neoplasms/immunology , Receptors, Immunologic/immunology , Toll-Like Receptors/immunology
17.
Cardiovasc Res ; 116(2): 295-305, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31150053

ABSTRACT

AIMS: The immune system is strongly involved in atherosclerosis and immune regulation generally leads to attenuated atherosclerosis. B- and T-lymphocyte attenuator (BTLA) is a novel co-receptor that negatively regulates the activation of B and T cells; however, there have been no reports of BTLA and its function in atherosclerosis or cardiovascular disease (CVD). We aimed to assess the dominant BTLA expressing leucocyte in CVD patients and to investigate whether BTLA has a functional role in experimental atherosclerosis. METHODS AND RESULTS: We show that BTLA is primarily expressed on B cells in CVD patients and follicular B2 cells in low-density lipoprotein receptor-deficient (Ldlr-/-) mice. We treated Ldlr-/- mice that were fed a western-type diet (WTD) with phosphate-buffered saline, an isotype antibody, or an agonistic BTLA antibody (3C10) for 6 weeks. We report here that the agonistic BTLA antibody significantly attenuated atherosclerosis. This was associated with a strong reduction in follicular B2 cells, while regulatory B and T cells were increased. The BTLA antibody showed similar immunomodulating effects in a progression study in which Ldlr-/- mice were fed a WTD for 10 weeks before receiving antibody treatment. Most importantly, BTLA stimulation enhanced collagen content, a feature of stable lesions, in pre-existing lesions. CONCLUSION: Stimulation of the BTLA pathway in Ldlr-/- mice reduces initial lesion development and increases collagen content of established lesions, presumably by shifting the balance between atherogenic follicular B cells and atheroprotective B cells and directing CD4+ T cells towards regulatory T cells. We provide the first evidence that BTLA is a very promising target for the treatment of atherosclerosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , B-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Plaque, Atherosclerotic , Receptors, Immunologic/agonists , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Cells, Cultured , Collagen/metabolism , Diet, High-Fat , Disease Models, Animal , Disease Progression , Humans , Male , Mice, Knockout , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
18.
J Med Chem ; 63(1): 309-320, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31809053

ABSTRACT

Mycobacterium tuberculosis (Mtb) continues to be a major health threat worldwide, and the development of Mtb vaccines could play a pivotal role in the prevention and control of this devastating epidemic. Th17-mediated immunity has been implicated in disease protection correlates of immune protection against Mtb. Currently, there are no approved adjuvants capable of driving a Th17 response in a vaccine setting. Recent clinical trial results using trehalose dibehenate have demonstrated a formulation-dependant proof of concept adjuvant system CAF01 capable of inducing long-lived protection. We have discovered a new class of Th17-inducing vaccine adjuvants based on the natural product Brartemicin. We synthesized and evaluated the capacity of a library of aryl trehalose derivatives to drive immunostimulatory reresponses and evaluated the structure-activity relationships in terms of the ability to engage the Mincle receptor and induce production of innate cytokines from human and murine cells. We elaborated on the structure-activity relationship of the new scaffold and demonstrated the ability of the lead entity to induce a pro-Th17 cytokine profile from primary human peripheral blood mononuclear cells and demonstrated efficacy in generating antibodies in combination with tuberculosis antigen M72 in a mouse model.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Mycobacterium tuberculosis/drug effects , Trehalose/analogs & derivatives , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/metabolism , Animals , Binding Sites , Cattle , Cell Line , Female , Humans , Lectins, C-Type/agonists , Lectins, C-Type/metabolism , Mice, Inbred BALB C , Molecular Docking Simulation , Molecular Structure , Receptors, Immunologic/agonists , Receptors, Immunologic/metabolism , Structure-Activity Relationship , Trehalose/chemical synthesis , Trehalose/metabolism , Trehalose/therapeutic use , Tuberculosis/therapy , Tuberculosis Vaccines/therapeutic use
19.
Org Biomol Chem ; 18(3): 425-430, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31774425

ABSTRACT

Herein, we report on the synthesis of a series of enantiomerically pure linear, iso-branched, and α-branched monoacyl glycerides (MAGs) in 63-72% overall yield. The ability of the MAGs to signal through human macrophage inducible C-type lectin (hMincle) using NFAT-GFP reporter cells was explored, as was the ability of the compounds to activate human monocytes. From these studies, MAGs with an acyl chain length ≥C22 were required for Mincle activation and the production of interleukin-8 (IL-8) by human monocytes. Moreover, the iso-branched MAGs led to a more pronounced immune response compared to linear MAGs, while an α-branched MAG containing a C-32 acyl chain activated cells to a higher degree than trehalose dibehenate (TDB), the prototypical Mincle agonist. Across the compound classes, the activity of the sn-1 substituted isomers was greater than the sn-3 counterparts. None of the representative compounds were cytotoxic, thus mitigating cytotoxicity as a potential mediator of cellular activity. Taken together, 6h (sn-1, iC26+1), 8a (sn-1, C32) and 8b (sn-3, C32) exhibited the best immunostimulatory properties and thus, have potential as vaccine adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Lectins, C-Type/agonists , Monoglycerides/pharmacology , Receptors, Immunologic/agonists , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/toxicity , Cell Line, Tumor , Humans , Molecular Structure , Monoglycerides/chemical synthesis , Monoglycerides/toxicity , Stereoisomerism , Structure-Activity Relationship
20.
Emerg Microbes Infect ; 8(1): 1168-1177, 2019.
Article in English | MEDLINE | ID: mdl-31379262

ABSTRACT

Mannose-capped lipoarabinomannan (ManLAM) is a high molecular mass amphipathic lipoglycan identified in pathogenic Mycobacterium tuberculosis (M. tb) and M. bovis Bacillus Calmette-Guérin (BCG). ManLAM, serves as both an immunogen and a modulator of the host immune system, and its critical role in mycobacterial survival during infection has been well-characterized. ManLAM can be recognized by various types of receptors on both innate and adaptive immune cells, including macrophages, dendritic cells (DCs), neutrophils, natural killer T (NKT) cells, T cells and B cells. MamLAM has been shown to affect phagocytosis, cytokine production, antigen presentation, T cell activation and polarization, as well as antibody production. Exploring the mechanisms underlying the roles of ManLAM during mycobacterial infection will aid in improving tuberculosis (TB) prevention, diagnosis and treatment interventions. In this review, we highlight the interaction between ManLAM and receptors, intracellular signalling pathways triggered by ManLAM and its roles in both innate and adaptive immune responses.


Subject(s)
Adaptive Immunity/drug effects , Immunity, Innate/drug effects , Immunologic Factors/analysis , Lipopolysaccharides/analysis , Mannose/analysis , Mycobacterium bovis/chemistry , Mycobacterium tuberculosis/chemistry , Animals , Humans , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Receptors, Immunologic/agonists , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...