Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(5): 101560, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38729159

ABSTRACT

Stimulator of IFN genes (STING) is a promising target for adjuvants utilized in in situ cancer vaccination approaches. However, key barriers remain for clinical translation, including low cellular uptake and accessibility, STING variability necessitating personalized STING agonists, and interferon (IFN)-independent signals that can promote tumor growth. Here, we identify C100, a highly deacetylated chitin-derived polymer (HDCP), as an attractive alternative to conventional STING agonists. C100 promotes potent anti-tumor immune responses, outperforming less deacetylated HDCPs, with therapeutic efficacy dependent on STING and IFN alpha/beta receptor (IFNAR) signaling and CD8+ T cell mediators. Additionally, C100 injection synergizes with systemic checkpoint blockade targeting PD-1. Mechanistically, C100 triggers mitochondrial stress and DNA damage to exclusively activate the IFN arm of the cGAS-STING signaling pathway and elicit sustained IFNAR signaling. Altogether, these results reveal an effective STING- and IFNAR-dependent adjuvant for in situ cancer vaccines with a defined mechanism and distinct properties that overcome common limitations of existing STING therapeutics.


Subject(s)
Adjuvants, Immunologic , CD8-Positive T-Lymphocytes , Chitin , Membrane Proteins , Mice, Inbred C57BL , Receptor, Interferon alpha-beta , Signal Transduction , Animals , Membrane Proteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Signal Transduction/drug effects , Humans , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Female , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Neoplasms/immunology , Neoplasms/therapy
2.
Cell Rep Med ; 4(1): 100899, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36652908

ABSTRACT

The non-canonical inflammasome sensor caspase-11 and gasdermin D (GSDMD) drive inflammation and pyroptosis, a type of immunogenic cell death that favors cell-mediated immunity (CMI) in cancer, infection, and autoimmunity. Here we show that caspase-11 and GSDMD are required for CD8+ and Th1 responses induced by nanoparticulate vaccine adjuvants. We demonstrate that nanoparticle-induced reactive oxygen species (ROS) are size dependent and essential for CMI, and we identify 50- to 60-nm nanoparticles as optimal inducers of ROS, GSDMD activation, and Th1 and CD8+ responses. We reveal a division of labor for IL-1 and IL-18, where IL-1 supports Th1 and IL-18 promotes CD8+ responses. Exploiting size as a key attribute, we demonstrate that biodegradable poly-lactic co-glycolic acid nanoparticles are potent CMI-inducing adjuvants. Our work implicates ROS and the non-canonical inflammasome in the mode of action of polymeric nanoparticulate adjuvants and establishes adjuvant size as a key design principle for vaccines against cancer and intracellular pathogens.


Subject(s)
Inflammasomes , Nanoparticles , Inflammasomes/metabolism , Interleukin-18/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Reactive Oxygen Species/metabolism , Phosphate-Binding Proteins/metabolism , Caspases/metabolism , Interleukin-1/metabolism
3.
Sci Immunol ; 7(78): eade5728, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36525507

ABSTRACT

Interleukin-1 (IL-1) family cytokines are key barrier cytokines that are typically expressed as inactive, or partially active, precursors that require proteolysis within their amino termini for activation. IL-37 is an enigmatic member of the IL-1 family that has been proposed to be activated by caspase-1 and to exert anti-inflammatory activity through engagement of the IL-18R and SIGIRR. However, here we show that the longest IL-37 isoform, IL-37b, exhibits robust proinflammatory activity upon amino-terminal proteolysis by neutrophil elastase or cathepsin S. In sharp contrast, caspase-1 failed to process or activate IL-37 at concentrations that robustly activated its canonical substrate, IL-1ß. IL-37 and IL-36 exhibit high structural homology, and, consistent with this, a K53-truncated form of IL-37, mimicking the cathepsin S-processed form of this cytokine, was found to exert its proinflammatory effects via IL-36 receptor engagement and produced an inflammatory signature practically identical to IL-36. Administration of K53-truncated IL-37b intraperitoneally into wild-type mice also elicited an inflammatory response that was attenuated in IL-36R-/- animals. These data demonstrate that, in common with other IL-1 family members, mature IL-37 can also elicit proinflammatory effects upon processing by specific proteases.


Subject(s)
Interleukin-1 , Peptide Hydrolases , Receptors, Interleukin , Animals , Mice , Caspases , Cathepsins , Cytokines , Interleukin-1/metabolism , Myeloid Cells , Receptors, Interleukin/metabolism
4.
PLoS One ; 16(8): e0244166, 2021.
Article in English | MEDLINE | ID: mdl-34347781

ABSTRACT

Fluorogenic aptamers are an alternative to established methodology for real-time imaging of RNA transport and dynamics. We developed Broccoli-aptamer concatemers ranging from 4 to 128 substrate-binding site repeats and characterized their behavior fused to an mCherry-coding mRNA in transient transfection, stable expression, and in recombinant cytomegalovirus infection. Concatemerization of substrate-binding sites increased Broccoli fluorescence up to a concatemer length of 16 copies, upon which fluorescence did not increase and mCherry signals declined. This was due to the combined effects of RNA aptamer aggregation and reduced RNA stability. Unfortunately, both cellular and cytomegalovirus genomes were unable to maintain and express high Broccoli concatemer copy numbers, possibly due to recombination events. Interestingly, negative effects of Broccoli concatemers could be partially rescued by introducing linker sequences in between Broccoli repeats warranting further studies. Finally, we show that even though substrate-bound Broccoli is easily photobleached, it can still be utilized in live-cell imaging by adapting a time-lapse imaging protocol.


Subject(s)
Brassica/genetics , RNA Stability/genetics , RNA, Messenger/genetics , Aptamers, Nucleotide/genetics , Brassica/virology , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/genetics , Fluorescence , Fluorescent Dyes/administration & dosage
5.
Biomaterials ; 275: 120961, 2021 08.
Article in English | MEDLINE | ID: mdl-34171753

ABSTRACT

Chitosan is a cationic polysaccharide that has been evaluated as an adjuvant due to its biocompatible and biodegradable nature. The polysaccharide can enhance antibody responses and cell-mediated immunity following vaccination by injection or mucosal routes. However, the optimal polymer characteristics for activation of dendritic cells (DCs) and induction of antigen-specific cellular immune responses have not been resolved. Here, we demonstrate that only chitin-derived polymers with a high degree of deacetylation (DDA) enhance generation of mitochondrial reactive oxygen species (mtROS), leading to cGAS-STING mediated induction of type I IFN. Additionally, the capacity of the polymers to activate the NLRP3 inflammasome was strictly dependent on the degree and pattern of deacetylation and mtROS generation. Polymers with a DDA below 80% are poor adjuvants while a fully deacetylated polyglucosamine polymer is most effective as a vaccine adjuvant. Furthermore, this polyglucosamine polymer enhanced antigen-specific Th1 responses in a NLRP3 and STING-type I IFN-dependent manner. Overall these results indicate that the degree of chitin deacetylation, the acetylation pattern and its regulation of mitochondrial ROS are the key determinants of its immune enhancing effects.


Subject(s)
Inflammasomes , Membrane Proteins , NLR Family, Pyrin Domain-Containing 3 Protein , Chitin , Mitochondria , Nucleotidyltransferases , Polymers , Reactive Oxygen Species
6.
Respir Res ; 22(1): 133, 2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33926483

ABSTRACT

Nutritional immunity is the sequestration of bioavailable trace metals such as iron, zinc and copper by the host to limit pathogenicity by invading microorganisms. As one of the most conserved activities of the innate immune system, limiting the availability of free trace metals by cells of the immune system serves not only to conceal these vital nutrients from invading bacteria but also operates to tightly regulate host immune cell responses and function. In the setting of chronic lung disease, the regulation of trace metals by the host is often disrupted, leading to the altered availability of these nutrients to commensal and invading opportunistic pathogenic microbes. Similarly, alterations in the uptake, secretion, turnover and redox activity of these vitally important metals has significant repercussions for immune cell function including the response to and resolution of infection. This review will discuss the intricate role of nutritional immunity in host immune cells of the lung and how changes in this fundamental process as a result of chronic lung disease may alter the airway microbiome, disease progression and the response to infection.


Subject(s)
Adaptive Immunity , Asthma/immunology , Communicable Diseases/immunology , Immunity, Innate , Lung/immunology , Metals/immunology , Microbiota , Nutritional Status , Pulmonary Disease, Chronic Obstructive/immunology , Animals , Asthma/microbiology , Asthma/physiopathology , Asthma/virology , Communicable Diseases/microbiology , Communicable Diseases/physiopathology , Communicable Diseases/virology , Host-Pathogen Interactions , Humans , Lung/microbiology , Lung/physiopathology , Lung/virology , Metals/metabolism , Prognosis , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/virology
7.
Int J Mol Sci ; 22(6)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33802220

ABSTRACT

Metabolic glycoengineering enables a directed modification of cell surfaces by introducing target molecules to surface proteins displaying new features. Biochemical pathways involving glycans differ in dependence on the cell type; therefore, this technique should be tailored for the best results. We characterized metabolic glycoengineering in telomerase-immortalized human mesenchymal stromal cells (hMSC-TERT) as a model for primary hMSC, to investigate its applicability in TERT-modified cell lines. The metabolic incorporation of N-azidoacetylmannosamine (Ac4ManNAz) and N-alkyneacetylmannosamine (Ac4ManNAl) into the glycocalyx as a first step in the glycoengineering process revealed no adverse effects on cell viability or gene expression, and the in vitro multipotency (osteogenic and adipogenic differentiation potential) was maintained under these adapted culture conditions. In the second step, glycoengineered cells were modified with fluorescent dyes using Cu-mediated click chemistry. In these analyses, the two mannose derivatives showed superior incorporation efficiencies compared to glucose and galactose isomers. In time-dependent experiments, the incorporation of Ac4ManNAz was detectable for up to six days while Ac4ManNAl-derived metabolites were absent after two days. Taken together, these findings demonstrate the successful metabolic glycoengineering of immortalized hMSC resulting in transient cell surface modifications, and thus present a useful model to address different scientific questions regarding glycosylation processes in skeletal precursors.


Subject(s)
Glycocalyx , Hexosamines , Mesenchymal Stem Cells/metabolism , Metabolic Engineering , Models, Biological , Myoblasts, Skeletal/metabolism , Cell Line, Transformed , Glycocalyx/chemistry , Glycocalyx/metabolism , Hexosamines/chemistry , Hexosamines/metabolism , Humans
8.
Semin Immunol ; 56: 101542, 2021 08.
Article in English | MEDLINE | ID: mdl-34973890

ABSTRACT

The dogma that immunological memory is an exclusive trait of adaptive immunity has been recently challenged by studies showing that priming of innate cells can also result in modified long-term responsiveness to secondary stimuli, once the cells have returned to a non-activated state. This phenomenon is known as 'innate immune memory', 'trained immunity' or 'innate training'. While the main known triggers of trained immunity are microbial-derived molecules such as ß-glucan, endogenous particles such as oxidized low-density lipoprotein and monosodium urate crystals can also induce trained phenotypes in innate cells. Whether exogenous particles can induce trained immunity has been overlooked. Our exposure to particulates has dramatically increased in recent decades as a result of the broad medical use of particle-based drug carriers, theragnostics, adjuvants, prosthetics and an increase in environmental pollution. We recently showed that pristine graphene can induce trained immunity in macrophages, enhancing their inflammatory response to TLR agonists, proving that exogenous nanomaterials can affect the long-term response of innate cells. The consequences of trained immunity can be beneficial, for instance, enhancing protection against unrelated pathogens; however, they can also be deleterious if they enhance inflammatory disorders. Therefore, studying the ability of particulates and biomaterials to induce innate trained phenotypes in cells is warranted. Here we analyse the mechanisms whereby particles can induce trained immunity and discuss how physicochemical characteristics of particulates could influence the induction of innate memory. We review the implications of trained immunity in the context of particulate adjuvants, nanocarriers and nanovaccines and their potential applications in medicine. Finally, we reflect on the unanswered questions and the future of the field.


Subject(s)
Immunity, Innate , Nanoparticles , Adaptive Immunity , Adjuvants, Immunologic , Humans , Immunologic Memory , Macrophages
9.
PLoS One ; 15(11): e0239713, 2020.
Article in English | MEDLINE | ID: mdl-33211703

ABSTRACT

Uncoupling proteins (UCPs) are members of the mitochondrial anion carrier superfamily that can mediate the transfer of protons into the mitochondrial matrix from the intermembrane space. We have previously reported UCP3 expression in thymocytes, mitochondria of total splenocytes and splenic lymphocytes. Here, we demonstrate that Ucp3 is expressed in peripheral naive CD4+ T cells at the mRNA level before being markedly downregulated following activation. Non-polarized, activated T cells (Th0 cells) from Ucp3-/- mice produced significantly more IL-2, had increased expression of CD25 and CD69 and were more proliferative than Ucp3+/+ Th0 cells. The altered IL-2 expression observed between T cells from Ucp3+/+ and Ucp3-/- mice may be a factor in determining differentiation into Th17 or induced regulatory (iTreg) cells. When compared to Ucp3+/+, CD4+ T cells from Ucp3-/- mice had increased FoxP3 expression under iTreg conditions. Conversely, Ucp3-/- CD4+ T cells produced a significantly lower concentration of IL-17A under Th17 cell-inducing conditions in vitro. These effects were mirrored in antigen-specific T cells from mice immunized with KLH and CT. Interestingly, the altered responses of Ucp3-/- T cells were partially reversed upon neutralisation of IL-2. Together, these data indicate that UCP3 acts to restrict the activation of naive T cells, acting as a rheostat to dampen signals following TCR and CD28 co-receptor ligation, thereby limiting early activation responses. The observation that Ucp3 ablation alters the Th17:Treg cell balance in vivo as well as in vitro suggests that UCP3 is a potential target for the treatment of Th17 cell-mediated autoimmune diseases.


Subject(s)
Lymphocyte Activation/genetics , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology , Uncoupling Protein 3/genetics , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Differentiation/immunology , Cell Proliferation/genetics , Interleukin-17/metabolism , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Uncoupling Protein 3/metabolism
10.
J Org Chem ; 85(15): 9751-9760, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32686416

ABSTRACT

A simple and efficient microwave-assisted synthesis of asymmetric pentamethine cyanine dyes with various functional groups was developed, which allows high-yielding results. The synthesized dyes are modifiable and suitable for single-molecule imaging in biological and medical sciences by application of click chemistry or classic esterification and amidation.

11.
Cell Rep ; 30(1): 124-136.e4, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914380

ABSTRACT

Increased glycolytic metabolism recently emerged as an essential process driving host defense against Mycobacterium tuberculosis (Mtb), but little is known about how this process is regulated during infection. Here, we observe repression of host glycolysis in Mtb-infected macrophages, which is dependent on sustained upregulation of anti-inflammatory microRNA-21 (miR-21) by proliferating mycobacteria. The dampening of glycolysis by miR-21 is mediated through targeting of phosphofructokinase muscle (PFK-M) isoform at the committed step of glycolysis, which facilitates bacterial growth by limiting pro-inflammatory mediators, chiefly interleukin-1ß (IL-1ß). Unlike other glycolytic genes, PFK-M expression and activity is repressed during Mtb infection through miR-21-mediated regulation, while other less-active isoenzymes dominate. Notably, interferon-γ (IFN-γ), which drives Mtb host defense, inhibits miR-21 expression, forcing an isoenzyme switch in the PFK complex, augmenting PFK-M expression and macrophage glycolysis. These findings place the targeting of PFK-M by miR-21 as a key node controlling macrophage immunometabolic function.


Subject(s)
Glycolysis , Host-Pathogen Interactions , Interleukin-1beta/metabolism , MicroRNAs/metabolism , Mycobacterium tuberculosis/physiology , Phosphofructokinase-1/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Base Sequence , Cell Proliferation , HEK293 Cells , Humans , Interferon-gamma/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , MicroRNAs/genetics , Phosphofructokinase-1/genetics , RAW 264.7 Cells , Tuberculosis/microbiology
12.
Vaccine ; 38(3): 635-643, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31727505

ABSTRACT

Despite being in the midst of a global pandemic of infections caused by the pathogen Chlamydia trachomatis, a vaccine capable of inducing protective immunity remains elusive. Given the C. trachomatis mucosal port of entry, a formulation compatible with mucosal administration and capable of eliciting potent genital tract immunity is highly desirable. While subunit vaccines are considered safer and better tolerated, these are typically poorly immunogenic and require co-formulation with immune-potentiating adjuvants. However, of the adjuvants licensed for use in humans, very few drive robust cellular responses, a pre-requisite for protection against C. trachomatis infection. Recently, the cationic adjuvant formulations (CAF) have been shown to induce robust humoral and cellular immunity in pre-clinical models of chlamydia, malaria and tuberculosis (TB). Here, we demonstrate that CAF01 induces potent immune responses when combined with the major outer membrane protein (MOMP) of C. trachomatis following parenteral immunisation and also as part of a heterologous prime/boost regime. We show that a subcutaneous prime with CAF01-adjuvanted recombinant MOMP licenses antigen-specific immunity at distant mucosal sites which can be activated following oral antigen re-encounter in the absence of concomitant adjuvant stimulation. Finally, we shed light on the mechanism(s) through which CAF01 elicits robust antigen-specific immunity to co-formulated MOMP via type I interferon (IFN) signalling.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Chlamydia trachomatis/drug effects , Immunity, Cellular/drug effects , Interferon Type I , Animals , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Drug Compounding/methods , Female , Immunity, Cellular/immunology , Interferon Type I/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/drug effects , Mucous Membrane/immunology
13.
Commun Biol ; 2: 261, 2019.
Article in English | MEDLINE | ID: mdl-31341960

ABSTRACT

Genetic code expansion (GCE) technology allows the specific incorporation of functionalized noncanonical amino acids (ncAAs) into proteins. Here, we investigated the Diels-Alder reaction between trans-cyclooct-2-ene (TCO)-modified ncAAs, and 22 known and novel 1,2,4,5-tetrazine-dye conjugates spanning the entire visible wavelength range. A hallmark of this reaction is its fluorogenicity - the tetrazine moiety can elicit substantial quenching of the dye. We discovered that photoinduced electron transfer (PET) from the excited dye to tetrazine is the main quenching mechanism in red-absorbing oxazine and rhodamine derivatives. Upon reaction with dienophiles quenching interactions are reduced resulting in a considerable increase in fluorescence intensity. Efficient and specific labeling of all tetrazine-dyes investigated permits super-resolution microscopy with high signal-to-noise ratio even at the single-molecule level. The different cell permeability of tetrazine-dyes can be used advantageously for specific intra- and extracellular labeling of proteins and highly sensitive fluorescence imaging experiments in fixed and living cells.


Subject(s)
Fluorescent Dyes/chemistry , Genetic Code , Microscopy, Confocal/instrumentation , Optical Imaging/instrumentation , Animals , COS Cells , Chlorocebus aethiops , Coloring Agents/chemistry , Cycloaddition Reaction , Cyclooctanes/chemistry , HEK293 Cells , Heterocyclic Compounds/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Microscopy, Confocal/methods , Optical Imaging/methods , Rhodamines , Staining and Labeling
15.
Immunity ; 50(6): 1412-1424.e6, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31076360

ABSTRACT

Assembly of inflammasomes after infection or injury leads to the release of interleukin-1ß (IL-1ß) and to pyroptosis. After inflammasome activation, cells either pyroptose or enter a hyperactivated state defined by IL-1ß secretion without cell death, but what controls these different outcomes is unknown. Here, we show that removal of the Toll-IL-1R protein SARM from macrophages uncouples inflammasome-dependent cytokine release and pyroptosis, whereby cells displayed increased IL-1ß production but reduced pyroptosis. Correspondingly, increasing SARM in cells caused less IL-1ß release and more pyroptosis. SARM suppressed IL-1ß by directly restraining the NLRP3 inflammasome and, hence, caspase-1 activation. Consistent with a role for SARM in pyroptosis, Sarm1-/- mice were protected from lipopolysaccharide (LPS)-stimulated sepsis. Pyroptosis-inducing, but not hyperactivating, NLRP3 stimulants caused SARM-dependent mitochondrial depolarization. Thus, SARM-dependent mitochondrial depolarization distinguishes NLRP3 activators that cause pyroptosis from those that do not, and SARM modulation represents a cell-intrinsic mechanism to regulate cell fate after inflammasome activation.


Subject(s)
Armadillo Domain Proteins/metabolism , Cytokines/metabolism , Cytoskeletal Proteins/metabolism , Inflammasomes/metabolism , Animals , Armadillo Domain Proteins/genetics , Biomarkers , Cell Survival , Cytoskeletal Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Binding , Pyroptosis , Signal Transduction
16.
J Immunol ; 202(4): 1145-1152, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30642984

ABSTRACT

The cytokine IL-33 is a well-established inducer of Th2 responses. However, roles for IL-33 in promoting CD8, Th1, and T regulatory cell responses have also emerged. In this study, the role of IL-33 as a regulator of particulate vaccine adjuvant-induced Ag-specific cellular immunity was investigated. We found that polymeric nanoparticles surpassed alum in their ability to enhance Ag-specific CD8 and Th1 responses. IL-33 was a potent negative regulator of both CD8+ T cell and Th1 responses following i.m. vaccination with Ag and nanoparticles, whereas the cytokine was required for the nanoparticle enhancement in Ag-specific IL-10. In contrast to the effect on cellular immunity, Ab responses were comparable between vaccinated wild-type and IL-33-deficient mice. IL-33 did not compromise alum-induced adaptive cellular immunity after i.m. vaccination. These data suggest that IL-33 attenuates the induction of cellular immune responses by nanoparticulate adjuvants and should be considered in the rational design of vaccines targeting enhanced CD8 and Th1 responses.


Subject(s)
Antigens/immunology , Immunity, Cellular/immunology , Interleukin-33/immunology , Vaccines/immunology , Animals , Antigens/administration & dosage , Injections, Intramuscular , Interleukin-33/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Vaccination , Vaccines/administration & dosage
17.
FEBS J ; 285(13): 2377-2401, 2018 07.
Article in English | MEDLINE | ID: mdl-29656546

ABSTRACT

Growing awareness of the multiplicity of roles for the IL-1 family in immune regulation has prompted research exploring these cytokines in the context of vaccine-induced immunity. While tightly regulated, cytokines of the IL-1 family are normally released in response to cellular stress and in combination with other danger-/damage-associated molecular patterns (DAMPs), triggering potent local and systemic immune responses. In the context of infection or autoimmunity, engagement of IL-1 family receptors links robust innate responses to adaptive immunity. Clinical and experimental evidence has revealed that many vaccine adjuvants induce the release of one or multiple IL-1 family cytokines. The coordinated release of IL-1 family members in response to adjuvant-induced damage or cell death may be a determining factor in the transition from local inflammation to the induction of an adaptive response. Here, we analyse the effects of IL-1 family cytokines on innate and adaptive immunity with a particular emphasis on activation of antigen-presenting cells and induction of T cell-mediated immunity, and we address in detail the contribution of these cytokines to the modes of action of vaccine adjuvants including those currently approved for human use.


Subject(s)
Adaptive Immunity/immunology , Cytokines/immunology , Immunity, Innate/immunology , Interleukin-1/immunology , Adaptive Immunity/drug effects , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cytokines/metabolism , Humans , Immunity, Innate/drug effects , Interleukin-1/metabolism , Stress, Physiological/drug effects , Stress, Physiological/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
18.
Eur J Immunol ; 48(4): 705-715, 2018 04.
Article in English | MEDLINE | ID: mdl-29349774

ABSTRACT

The effectiveness of many vaccines licensed for clinical use relates to the induction of neutralising antibodies, facilitated by the inclusion of vaccine adjuvants, particularly alum. However, the ability of alum to preferentially promote humoral rather than cellular, particularly Th1-type responses, is not well understood. We demonstrate that alum activates immunosuppressive mechanisms following vaccination, which limit its capacity to induce Th1 responses. One of the key cytokines limiting excessive immune responses is IL-10. Injection of alum primed draining lymph node cells for enhanced IL-10 secretion ex vivo. Moreover, at the site of injection, macrophages and dendritic cells were key sources of IL-10 expression. Alum strongly enhanced the transcription and secretion of IL-10 by macrophages and dendritic cells. The absence of IL-10 signalling did not compromise alum-induced cell infiltration into the site of injection, but resulted in enhanced antigen-specific Th1 responses after vaccination. In contrast to its decisive regulatory role in regulating Th1 responses, there was no significant change in antigen-specific IgG1 antibody production following vaccination with alum in IL-10-deficient mice. Overall, these findings indicate that injection of alum promotes IL-10, which can block Th1 responses and may explain the poor efficacy of alum as an adjuvant for inducing protective Th1 immunity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Dendritic Cells/immunology , Interleukin-10/immunology , Macrophages/immunology , Monocytes/immunology , Th1 Cells/immunology , Animals , Cells, Cultured , Escherichia coli/immunology , Female , Interleukin-10/biosynthesis , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccines/immunology
19.
Chemistry ; 23(48): 11684-11693, 2017 Aug 25.
Article in English | MEDLINE | ID: mdl-28631852

ABSTRACT

Salts of anionic silver(I) clusters with the carba-closo-dodecaboranylethynyl ligand were obtained from {Ag2 (12-C≡C-closo-1-CB11 H11 )}n , selected pyridines, and [Et4 N]Cl or [Ph4 P]Br. Salts of octahedral silver(I) clusters [Et4 N]2 [Ag6 (12-C≡C-closo-1-CB11 H11 )4 (4-X-C5 H5 N)x ] were formed with pyridine (X=H, x=8), 4-methylpyridine (X=Me, x=8), and 4-cyanopyridine (X=CN, x=10). In contrast, 3,5-lutidine (3,5-Me2 Py) did not result in salts of dianionic clusters, even in the presence of excess of [Et4 N]Cl or [Ph4 P]Br; instead salts of monoanionic AgI7 clusters, [Et4 N][Ag7 (12-C≡C-closo-1-CB11 H11 )4 (3,5-Me2 Py)9 ] and [Ph4 P][Ag7 (12-C≡C-closo-1-CB11 H11 )4 (3,5-Me2 Py)13 ] were obtained. The AgI7 cluster is pentagonal bipyramidal in the former, but is an edge-capped octahedron in the latter. The 4-methylpyridine and 3,5-lutidine complexes show green phosphorescence at room temperature. Although argentophilic interactions give rise to sufficient spin-orbit coupling for intersystem crossing S1 →Tn and moderate-to-high radiative rate constants, time-resolved measurements indicate that the quantum yields are greatly influenced by the pyridine ligands, which mainly determine the non-radiative rate constants. In addition, the crystal structures of [Ag16 (12-C≡C-closo-1-CB11 H11 )8 (Py)9.25 (CH3 CN)2 (CH2 Cl2 )0.75 ]⋅CH2 Cl2 , [Ag8 (12-C≡C-closo-1-CB11 H11 )4 (Py)12 ], [Ag10 (12-C≡C-closo-1-CB11 H11 )4 (4-MePy)10 Br2 ], [Ag7 (12-C≡C-closo-1-CB11 H11 )3 (4-tBuPy)11 Cl]⋅(4-tBuPy), and [Ag9 (12-C≡C-closo-1-CB11 H11 )4 (3,5-Me2 Py)11 Cl] were elucidated.

20.
Nat Cell Biol ; 19(4): 267-269, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28361938

ABSTRACT

The Hippo pathway responds to environmental factors including nutrient availability, cell density and substrate stiffness to regulate organ size. This pathway is now shown to also regulate antiviral defence by modulating the TBK1-mediated control of interferon production.


Subject(s)
Protein Serine-Threonine Kinases , Signal Transduction , Antiviral Agents , Organ Size
SELECTION OF CITATIONS
SEARCH DETAIL
...