Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep Med ; 5(5): 101560, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38729159

RESUMO

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.


Assuntos
Adjuvantes Imunológicos , Linfócitos T CD8-Positivos , Quitina , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta , Transdução de Sinais , Animais , Proteínas de Membrana/metabolismo , Proteínas de Membrana/imunologia , Proteínas de Membrana/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Receptor de Interferon alfa e beta/genética , Camundongos , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Humanos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Feminino , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Neoplasias/imunologia , Neoplasias/terapia
2.
Eur J Immunol ; 54(2): e2350512, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37994660

RESUMO

Vaccination is considered one of the major milestones in modern medicine, facilitating the control and eradication of life-threatening infectious diseases. Vaccine adjuvants are a key component of many vaccines, serving to steer antigen-specific immune responses and increase their magnitude. Despite major advances in the field of adjuvant research over recent decades, our understanding of their mechanism of action remains incomplete. This hinders our capacity to further improve these adjuvant technologies, so addressing how adjuvants induce and control the induction of innate and adaptive immunity is a priority. Investigating how adjuvant physicochemical properties, such as size and charge, exert immunomodulatory effects can provide valuable insights and serve as the foundation for the rational design of vaccine adjuvants. Most clinically applied adjuvants are particulate in nature and polymeric particulate adjuvants present advantages due to stability, biocompatibility profiles, and flexibility in terms of formulation. These properties can impact on antigen release kinetics and biodistribution, cellular uptake and targeting, and drainage to the lymphatics, consequently dictating the induction of innate, cellular, and humoral adaptive immunity. A current focus is to apply rational design principles to the development of adjuvants capable of eliciting robust cellular immune responses including CD8+ cytotoxic T-cell and Th1-biased CD4+ T-cell responses, which are required for vaccines against intracellular pathogens and cancer. This review highlights recent advances in our understanding of how particulate adjuvants, especially polymer-based particulates, modulate immune responses and how this can be used as a guide for improved adjuvant design.


Assuntos
Adjuvantes de Vacinas , Vacinas , Distribuição Tecidual , Vacinação , Imunidade Adaptativa , Adjuvantes Imunológicos/farmacologia , Antígenos
3.
Cell Rep Med ; 4(1): 100899, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36652908

RESUMO

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.


Assuntos
Inflamassomos , Nanopartículas , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Caspases/metabolismo , Interleucina-1/metabolismo
4.
Elife ; 112022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-36173104

RESUMO

Macrophages are a highly adaptive population of innate immune cells. Polarization with IFNγ and LPS into the 'classically activated' M1 macrophage enhances pro-inflammatory and microbicidal responses, important for eradicating bacteria such as Mycobacterium tuberculosis. By contrast, 'alternatively activated' M2 macrophages, polarized with IL-4, oppose bactericidal mechanisms and allow mycobacterial growth. These activation states are accompanied by distinct metabolic profiles, where M1 macrophages favor near exclusive use of glycolysis, whereas M2 macrophages up-regulate oxidative phosphorylation (OXPHOS). Here, we demonstrate that activation with IL-4 and IL-13 counterintuitively induces protective innate memory against mycobacterial challenge. In human and murine models, prior activation with IL-4/13 enhances pro-inflammatory cytokine secretion in response to a secondary stimulation with mycobacterial ligands. In our murine model, enhanced killing capacity is also demonstrated. Despite this switch in phenotype, IL-4/13 trained murine macrophages do not demonstrate M1-typical metabolism, instead retaining heightened use of OXPHOS. Moreover, inhibition of OXPHOS with oligomycin, 2-deoxy glucose or BPTES all impeded heightened pro-inflammatory cytokine responses from IL-4/13 trained macrophages. Lastly, this work identifies that IL-10 attenuates protective IL-4/13 training, impeding pro-inflammatory and bactericidal mechanisms. In summary, this work provides new and unexpected insight into alternative macrophage activation states in the context of mycobacterial infection.


Assuntos
Interleucina-10 , Interleucina-13 , Animais , Citocinas/metabolismo , Glucose/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Lipopolissacarídeos/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos , Oligomicinas , Fosforilação Oxidativa
5.
Nat Rev Immunol ; 22(4): 236-250, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34312520

RESUMO

Mucosal vaccines offer the potential to trigger robust protective immune responses at the predominant sites of pathogen infection. In principle, the induction of adaptive immunity at mucosal sites, involving secretory antibody responses and tissue-resident T cells, has the capacity to prevent an infection from becoming established in the first place, rather than only curtailing infection and protecting against the development of disease symptoms. Although numerous effective mucosal vaccines are in use, the major advances seen with injectable vaccines (including adjuvanted subunit antigens, RNA and DNA vaccines) have not yet been translated into licensed mucosal vaccines, which currently comprise solely live attenuated and inactivated whole-cell preparations. The identification of safe and effective mucosal adjuvants allied to innovative antigen discovery and delivery strategies is key to advancing mucosal vaccines. Significant progress has been made in resolving the mechanisms that regulate innate and adaptive mucosal immunity and in understanding the crosstalk between mucosal sites, and this provides valuable pointers to inform mucosal adjuvant design. In particular, increased knowledge on mucosal antigen-presenting cells, innate lymphoid cell populations and resident memory cells at mucosal sites highlights attractive targets for vaccine design. Exploiting these insights will allow new vaccine technologies to be leveraged to facilitate rational mucosal vaccine design for pathogens including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and for cancer.


Assuntos
COVID-19 , Vacinas , Adjuvantes Imunológicos , COVID-19/prevenção & controle , Humanos , Imunidade Inata , Imunidade nas Mucosas , Linfócitos , SARS-CoV-2
6.
Nat Commun ; 12(1): 1460, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33674584

RESUMO

Mitochondria are important regulators of macrophage polarisation. Here, we show that arginase-2 (Arg2) is a microRNA-155 (miR-155) and interleukin-10 (IL-10) regulated protein localized at the mitochondria in inflammatory macrophages, and is critical for IL-10-induced modulation of mitochondrial dynamics and oxidative respiration. Mechanistically, the catalytic activity and presence of Arg2 at the mitochondria is crucial for oxidative phosphorylation. We further show that Arg2 mediates this process by increasing the activity of complex II (succinate dehydrogenase). Moreover, Arg2 is essential for IL-10-mediated downregulation of the inflammatory mediators succinate, hypoxia inducible factor 1α (HIF-1α) and IL-1ß in vitro. Accordingly, HIF-1α and IL-1ß are highly expressed in an LPS-induced in vivo model of acute inflammation using Arg2-/- mice. These findings shed light on a new arm of IL-10-mediated metabolic regulation, working to resolve the inflammatory status of the cell.


Assuntos
Arginase/metabolismo , Interleucina-10/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Animais , Arginase/genética , Regulação para Baixo , Feminino , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Mitocôndrias/enzimologia , Succinato Desidrogenase/metabolismo
7.
Semin Immunol ; 56: 101542, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34973890

RESUMO

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.


Assuntos
Imunidade Inata , Nanopartículas , Imunidade Adaptativa , Adjuvantes Imunológicos , Humanos , Memória Imunológica , Macrófagos
8.
ACS Chem Biol ; 15(9): 2415-2421, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32786261

RESUMO

Macrophages are key immune cells for combatting Mycobacterium tuberculosis. However, M. tuberculosis possesses means to evade macrophage bactericidal responses by, for instance, secretion of the immunomodulatory para-hydroxybenzoic acid derivatives (pHBADs). While these molecules have been implicated in inhibiting macrophage responses in an acute context, little is known about their ability to reprogram macrophages via induction of long-term innate memory. Since innate memory has been highlighted as a promising strategy to augment bactericidal immune responses against M. tuberculosis, investigating corresponding immune evasion mechanisms is highly relevant. Our results reveal for the first time that pHBAD I and related molecules (unmethylated pHBAD I and the hexose l-rhamnose) reduce macrophage bactericidal mechanisms in both the short- and the long-term. Moreover, we demonstrate how methyl-p-anisate hinders bactericidal responses soon after exposure yet results in enhanced pro-inflammatory responses in the long-term. This work highlights new roles for these compounds in M. tuberculosis pathogenesis.


Assuntos
Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Parabenos/farmacologia , Animais , Benzoatos/farmacologia , Interleucina-10/metabolismo , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ramnose/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
9.
Immunol Rev ; 296(1): 169-190, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32594569

RESUMO

Therapeutic vaccination offers great promise as an intervention for a diversity of infectious and non-infectious conditions. Given that most chronic health conditions are thought to have an immune component, vaccination can at least in principle be proposed as a therapeutic strategy. Understanding the nature of protective immunity is of vital importance, and the progress made in recent years in defining the nature of pathological and protective immunity for a range of diseases has provided an impetus to devise strategies to promote such responses in a targeted manner. However, in many cases, limited progress has been made in clinical adoption of such approaches. This in part results from a lack of safe and effective vaccine adjuvants that can be used to promote protective immunity and/or reduce deleterious immune responses. Although somewhat simplistic, it is possible to divide therapeutic vaccine approaches into those targeting conditions where antibody responses can mediate protection and those where the principal focus is the promotion of effector and memory cellular immunity or the reduction of damaging cellular immune responses as in the case of autoimmune diseases. Clearly, in all cases of antigen-specific immunotherapy, the identification of protective antigens is a vital first step. There are many challenges to developing therapeutic vaccines beyond those associated with prophylactic diseases including the ongoing immune responses in patients, patient heterogeneity, and diversity in the type and stage of disease. If reproducible biomarkers can be defined, these could allow earlier diagnosis and intervention and likely increase therapeutic vaccine efficacy. Current immunomodulatory approaches related to adoptive cell transfers or passive antibody therapy are showing great promise, but these are outside the scope of this review which will focus on the potential for adjuvanted therapeutic active vaccination strategies.


Assuntos
Adjuvantes Imunológicos , Imunomodulação , Vacinação , Vacinas/imunologia , Vacinas/uso terapêutico , Animais , Formação de Anticorpos/imunologia , Autoimunidade , Gerenciamento Clínico , Humanos , Imunidade Celular , Imunidade Humoral , Terapia de Alvo Molecular , Resultado do Tratamento , Vacinação/métodos , Vacinas/administração & dosagem
10.
Nanoscale ; 12(20): 11192-11200, 2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32407430

RESUMO

Graphene-based materials are of increasing interest for their potential use in biomedical applications. However, there is a need to gain a deeper understanding of how graphene modulates biological responses before moving towards clinical application. Innate immune training is a recently described phenomenon whereby cells of the innate immune system are capable of being programmed to generate an increased non-specific response upon subsequent challenge. This has been well established in the case of certain microbes and microbial products. However, little is known about the capacity of particulate materials, such as pristine graphene (pGr), to promote innate immune training. Here we report for the first time that while stimulation with pGr alone does not directly induce cytokine secretion by bone-marrow derived macrophages (BMDMs), it programs them for enhanced secretion of proinflammatory cytokines (IL-6, TNF-α) and a concomitant decrease in production of the regulatory cytokine, IL-10 after Toll-like receptor (TLR) ligand stimulation. This capacity of pGr to program cells for enhanced inflammatory responses could be overcome if the nanomaterial is incorporated in a collagen matrix. Our findings thus demonstrate the potential of graphene to modulate innate immunity over long timescales and have implications for the design and biomedical use of pGr-based materials.


Assuntos
Fulerenos/farmacologia , Imunidade Inata/efeitos dos fármacos , Macrófagos/imunologia , Monocinas/imunologia , Receptores Toll-Like/imunologia , Animais , Fulerenos/química , Macrófagos/citologia , Camundongos
11.
Nat Commun ; 11(1): 1055, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-32103022

RESUMO

Activated caspase-1 and caspase-11 induce inflammatory cell death in a process termed pyroptosis. Here we show that Prostaglandin E2 (PGE2) inhibits caspase-11-dependent pyroptosis in murine and human macrophages. PGE2 suppreses caspase-11 expression in murine and human macrophages and in the airways of mice with allergic inflammation. Remarkably, caspase-11-deficient mice are strongly resistant to developing experimental allergic airway inflammation, where PGE2 is known to be protective. Expression of caspase-11 is elevated in the lung of wild type mice with allergic airway inflammation. Blocking PGE2 production with indomethacin enhances, whereas the prostaglandin E1 analog misoprostol inhibits lung caspase-11 expression. Finally, alveolar macrophages from asthma patients exhibit increased expression of caspase-4, a human homologue of caspase-11. Our findings identify PGE2 as a negative regulator of caspase-11-driven pyroptosis and implicate caspase-4/11 as a critical contributor to allergic airway inflammation, with implications for pathophysiology of asthma.


Assuntos
Asma/patologia , Caspases Iniciadoras/metabolismo , Dinoprostona/metabolismo , Macrófagos/imunologia , Piroptose/fisiologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Asma/imunologia , Caspases Iniciadoras/genética , Caspases Iniciadoras/imunologia , Células Cultivadas , Sinergismo Farmacológico , Feminino , Humanos , Indometacina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Misoprostol/farmacologia
12.
Cell Rep ; 30(1): 124-136.e4, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31914380

RESUMO

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.


Assuntos
Glicólise , Interações Hospedeiro-Patógeno , Interleucina-1beta/metabolismo , MicroRNAs/metabolismo , Mycobacterium tuberculosis/fisiologia , Fosfofrutoquinase-1/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Sequência de Bases , Proliferação de Células , Células HEK293 , Humanos , Interferon gama/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , MicroRNAs/genética , Fosfofrutoquinase-1/genética , Células RAW 264.7 , Tuberculose/microbiologia
13.
J Allergy Clin Immunol ; 145(4): 1272-1284.e6, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31983527

RESUMO

BACKGROUND: The World Health Organization estimates that air pollution is responsible for 7 million deaths per annum, with 7% of these attributable to pneumonia. Many of these fatalities have been linked to exposure to high levels of airborne particulates, such as diesel exhaust particles (DEPs). OBJECTIVES: We sought to determine whether exposure to DEPs could promote the progression of asymptomatic nasopharyngeal carriage of Streptococcus pneumoniae to invasive pneumococcal disease. METHODS: We used mouse models and in vitro assays to provide a mechanistic understanding of the link between DEP exposure and pneumococcal disease risk, and we confirmed our findings by using induced sputum macrophages isolated from healthy human volunteers. RESULTS: We demonstrate that inhaled exposure to DEPs disrupts asymptomatic nasopharyngeal carriage of S pneumoniae in mice, leading to dissemination to lungs and blood. Pneumococci are transported from the nasopharynx to the lungs following exposure to DEPs, leading to increased proinflammatory cytokine production, reduced phagocytic function of alveolar macrophages, and consequently, increased pneumococcal loads within the lungs and translocation into blood. These findings were confirmed by using DEP-exposed induced sputum macrophages isolated from healthy volunteers, demonstrating that impaired innate immune mechanisms following DEP exposure are also at play in humans. CONCLUSION: Lung inhaled DEPs increase susceptibility to pneumococcal disease by leading to loss of immunological control of pneumococcal colonisation, increased inflammation, tissue damage, and systemic bacterial dissemination.


Assuntos
Pulmão/imunologia , Macrófagos/imunologia , Nasofaringe/patologia , Material Particulado/efeitos adversos , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/fisiologia , Animais , Bacteriemia , Portador Sadio , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Humanos , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nasofaringe/microbiologia , Fagocitose , Pneumonia Pneumocócica/epidemiologia , Risco , Emissões de Veículos
14.
NPJ Vaccines ; 4: 45, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31666991

RESUMO

Helicobacter pylori causes chronic gastric infection that can lead to peptic ulcers and is an identified risk factor for gastric cancer development. Although much effort has been put into the development of a Helicobacter pylori vaccine over the last three decades, none has yet reached clinical application. Specific challenges pertaining to effective H. pylori vaccine development include the lack of proven vaccine-effective antigens and safe mucosal adjuvants to enhance local immune responses as well as the lack of accepted correlates of protection. Herein, we demonstrate that prophylactic intragastric immunisation with a whole-cell killed H. pylori antigen administered together with the non-toxic oral adjuvant α-galactosylceramide (α-GalCer) induced effective immune protection against H. pylori infection in mice, which was of similar magnitude as when using the "gold standard" cholera toxin as adjuvant. We further describe that this α-GalCer-adjuvanted vaccine formulation elicited strong intestinal and systemic Th1 responses as well as significant antigen-specific mucosal and systemic antibody responses. Finally, we report that the protective intestinal Th1 responses induced by α-GalCer are dependent on CD1d, IL-1R as well as IL-17R signalling. In summary, our results show that α-GalCer is a promising adjuvant for inclusion in an oral vaccine against H. pylori infection.

15.
Immunity ; 50(6): 1412-1424.e6, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31076360

RESUMO

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.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Citocinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Inflamassomos/metabolismo , Animais , Proteínas do Domínio Armadillo/genética , Biomarcadores , Sobrevivência Celular , Proteínas do Citoesqueleto/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ligação Proteica , Piroptose , Transdução de Sinais
16.
Biochem Pharmacol ; 163: 472-480, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30880061

RESUMO

Cyclosporine A has long been known to suppress T cell responses by inhibiting the production of IL-2, which drives T cell proliferation, enabling its use as a therapeutic for transplantation or autoimmunity. However, cyclosporine A also impacts on innate immune cells including dendritic cells, macrophages and neutrophils. In dendritic cells, which are essential for T cell priming, cyclosporine A can modulate both expression of surface molecules that engage with T cells and cytokine secretion, leading to altered induction of T cell responses. In macrophages and neutrophils, which play key antimicrobial roles, cyclosporine A reduces the production of cytokines that can play protective roles against pathogens. Some of these molecules, if produced in the context of chronic disease, can also contribute to pathology. There have been a number of elegant recent studies addressing the mechanisms by which cyclosporine A can modulate innate immunity. In particular, cyclosporine A inhibits the release of mitochondrial factors that stimulate the production of type 1 interferons by innate immune cells. This review addresses the emerging literature on modulation of innate immune responses by cyclosporine A, its resultant impact on adaptive immune responses and how this offers potential for new therapeutic applications.


Assuntos
Ciclosporina/farmacologia , Células Dendríticas/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Humanos , Neutrófilos/efeitos dos fármacos
17.
J Pharm Pharmacol ; 71(6): 920-928, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30805935

RESUMO

OBJECTIVES: The cationic biopolymer chitosan (CH) has emerged as a promising candidate adjuvant due to its safety profile and immunostimulatory properties. The presence of endotoxin contamination in biomaterials is generally underappreciated and can generate misleading results. It is important to establish a convenient methodology to obtain large amounts of high quality chitosan nanoparticles for biomedical applications. METHODS: We developed an easy method to generate endotoxin-free chitosan and assessed its purity using the Limulus amebocyte lysate assay and by measuring dendritic cell activation. KEY FINDINGS: Purified chitosan-based formulations alone failed to induce production of the proinflammatory cytokines tumour necrosis factor alpha (TNF-α) and interleukin (IL)-6 in bone marrow-derived dendritic cells (BMDCs) generated from C57BL/6 mice, while maintaining its ability to promote IL-1ß secretion in combination with the Toll-like receptor (TLR)-9 agonist, CpG. Moreover, BMDCs from C3H/HeN and TLR4-deficient mice, C3H/HeJ were stimulated with endotoxin-free chitosan-based formulations and no differences were observed in IL-6 and IL-1ß secretion, excluding the involvement of TLR-4 in the immunomodulatory effects of chitosan. CONCLUSIONS: The developed method provides simple guidelines for the production of endotoxin-free chitosan, ideal for biomedical applications.


Assuntos
Quitosana/farmacologia , Células Dendríticas/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Nanopartículas , Animais , Células da Medula Óssea/citologia , Quitosana/normas , Citocinas/metabolismo , Células Dendríticas/imunologia , Endotoxinas/análise , Feminino , Fatores Imunológicos/normas , Mediadores da Inflamação/metabolismo , Teste do Limulus/métodos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/farmacologia
18.
Int J Pharm ; 552(1-2): 7-15, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30244149

RESUMO

The use of tailored particle-based adjuvants constitutes a promising way to enhance antigen-specific humoral and cellular immune responses. However, a thorough understanding of the mechanisms underlying their adjuvanticity is crucial to generate more effective vaccines. We studied the ability of chitosan-aluminum nanoparticles (CH-Al NPs), which combine the immunostimulatory effects of chitosan and aluminum salts, to promote dendritic cell activation, assess their impact on innate and adaptive immune responses, and compare the results to those reported for conventional chitosan particles (CH-Na NPs). All tested CH-NP formulations were capable of modulating cytokine secretion by dendritic cells. CH-Al NPs promoted NLRP3 inflammasome activation, enhancing the release of IL-1ß without significantly inhibiting Th1 and Th17 cell-polarizing cytokines, IL-12p70 or IL-23, and induced DC maturation, but did not promote pro-inflammatory cytokine production on their own. In vivo results showed that mice injected with CH-Al NPs generated a local inflammatory response comparable to that elicited by the vaccine adjuvant alum. Importantly, after subcutaneous immunization with CH-Al NPs combined with the hepatitis B surface antigen (HBsAg), mice developed antigen-specific IgG titers in serum, nasal and vaginal washes. Overall, our results established CH-Al NPs as a potential adjuvant to enhance both innate and adaptive immune responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Alumínio/administração & dosagem , Quitosana/administração & dosagem , Antígenos de Superfície da Hepatite B/administração & dosagem , Nanopartículas/administração & dosagem , Animais , Citocinas/imunologia , Feminino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
20.
Eur J Immunol ; 48(4): 705-715, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29349774

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alúmen/farmacologia , Células Dendríticas/imunologia , Interleucina-10/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Células Th1/imunologia , Animais , Células Cultivadas , Escherichia coli/imunologia , Feminino , Interleucina-10/biossíntese , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vacinas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA