Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Nutrients ; 15(2)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36678282

ABSTRACT

Nutraceuticals act as cellular and functional modulators, contributing to the homeostasis of physiological processes. In an inflammatory microenvironment, these functional foods can interact with the immune system by modulating or balancing the exacerbated proinflammatory response. In this process, immune cells, such as antigen-presenting cells (APCs), identify danger signals and, after interacting with T lymphocytes, induce a specific effector response. Moreover, this conditions their change of state with phenotypical and functional modifications from the resting state to the activated and effector state, supposing an increase in their energy requirements that affect their intracellular metabolism, with each immune cell showing a unique metabolic signature. Thus, nutraceuticals, such as polyphenols, vitamins, fatty acids, and sulforaphane, represent an active option to use therapeutically for health or the prevention of different pathologies, including obesity, metabolic syndrome, and diabetes. To regulate the inflammation associated with these pathologies, intervention in metabolic pathways through the modulation of metabolic energy with nutraceuticals is an attractive strategy that allows inducing important changes in cellular properties. Thus, we provide an overview of the link between metabolism, immune function, and nutraceuticals in chronic inflammatory processes associated with obesity and diabetes, paying particular attention to nutritional effects on APC and T cell immunometabolism, as well as the mechanisms required in the change in energetic pathways involved after their activation.


Subject(s)
Antigen-Presenting Cells , T-Lymphocytes , Humans , Antigen-Presenting Cells/metabolism , Macrophages/metabolism , Inflammation/metabolism , Dietary Supplements , Obesity/metabolism
2.
Front Immunol ; 12: 687367, 2021.
Article in English | MEDLINE | ID: mdl-34394081

ABSTRACT

The essential microelement zinc plays immunoregulatory roles via its ability to influence signaling pathways. Zinc deficiency impairs overall immune function and resultantly increases susceptibility to infection. Thus, zinc is considered as an immune-boosting supplement for populations with hypozincemia at high-risk for infection. Besides its role as a structural cofactor of many proteins, zinc also acts as an intracellular messenger in immune cell signaling. T-cell activation instructs zinc influx from extracellular and subcellular sources through the Zip6 and Zip8 zinc transporters, respectively. Increased cytoplasmic zinc participates in the regulation of T-cell responses by modifying activation signaling. However, the mechanism underlying the activation-dependent movement of zinc ions by Zip transporters in T cells remains elusive. Here, we demonstrate that Zip6, one of the most abundantly expressed Zip transporters in T cells, is mainly localized to lipid rafts in human T cells and is recruited into the immunological synapse in response to TCR stimulation. This was demonstrated through confocal imaging of the interaction between CD4+ T cells and antigen-presenting cells. Further, immunoprecipitation assays show that TCR triggering induces tyrosine phosphorylation of Zip6, which has at least three putative tyrosine motifs in its long cytoplasmic region, and this phosphorylation is coupled with its physical interaction with Zap70. Silencing Zip6 reduces zinc influx from extracellular sources and suppresses T-cell responses, suggesting an interaction between Zip6-mediated zinc influx and TCR activation. These results provide new insights into the mechanism through which Zip6-mediated zinc influx occurs in a TCR activation-dependent manner in human CD4+ T cells.


Subject(s)
Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cation Transport Proteins/metabolism , Immunological Synapses/metabolism , Membrane Microdomains/metabolism , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Cation Transport Proteins/genetics , Humans , Immunological Synapses/immunology , Jurkat Cells , Lymphocyte Activation , Membrane Microdomains/immunology , Neoplasm Proteins/genetics , Phosphorylation , Signal Transduction , Tyrosine
3.
Cell Signal ; 83: 109994, 2021 07.
Article in English | MEDLINE | ID: mdl-33781846

ABSTRACT

Cells can communicate through the extracellular vesicles (EVs) they secrete. Pathogen associated molecular patterns (PAMPs), alter the biophysical and communicative properties of EVs released from cells, but the functional consequences of these changes are unknown. Characterization of keratinocyte-derived EVs after poly(I:C) treatment (poly(I:C)-EVs) showed slight differences in levels of EV markers TSG101 and Alix, a loss of CD63 and were positive for autophagosome marker LC3b-II and the cytokine IL36γ compared to EVs from unstimulated keratinocytes (control-EVs). Flagellin treatment (flagellin-EVs) led to an EV marker profile like control-EVs but lacked LC3b-II. Flagellin-EVs also lacked IL-36γ despite nearly identical intracellular levels. While poly(I:C) treatment led to the clear emergence of a > 200 nm diameter EV sub-population, these were not found in flagellin-EVs. EV associated IL-36γ colocalized with LC3b-II in density gradient analysis, equilibrating to 1.10 g/mL, indicating a common EV species. Poly(I:C), but not flagellin, induced intracellular vesicles positive for IL-36γ, LC3b-II, Alix and TSG101, consistent with fusion of autophagosomes and multivesicular bodies. Simultaneous rapamycin and flagellin treatment induced similar intracellular vesicles but was insufficient for the release of IL-36γ+/LC3b-II+ EVs. Finally, a qRT-PCR array screen showed eight cytokine/chemokine transcripts were altered (p < 0.05) in monocyte-derived Langerhans cells (LCs) when stimulated with poly(I:C)-EVs while three were altered when LCs were stimulated with flagellin-EVs compared to control-EVs. After independent confirmation, poly(I:C)-EVs upregulated BMP6 (p = 0.035) and flagellin-EVs upregulated CXCL8 (p = 0.005), VEGFA (p = 0.018) and PTGS2 (p = 0.020) compared to control-EVs. We conclude that exogenous signals derived from pathogens can alter keratinocyte-mediated modulation of the local immune responses by inducing changes in the types of EVs secreted and responses in antigen presenting cells.


Subject(s)
Antigen-Presenting Cells/metabolism , Extracellular Vesicles/metabolism , Keratinocytes/metabolism , Poly I-C/pharmacology , Toll-Like Receptors/agonists , Ascorbic Acid , Cells, Cultured , Cholecalciferol , Dehydroepiandrosterone/analogs & derivatives , Nicotinic Acids , Plant Extracts , Toll-Like Receptors/metabolism
4.
ACS Appl Mater Interfaces ; 12(49): 54399-54414, 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33215918

ABSTRACT

Tumor vaccines based on synthetic human papillomavirus (HPV) oncoprotein E7 and/or E6 peptides have shown encouraging results in preclinical model studies and human clinical trials. However, the clinical efficacy may be limited by the disadvantages of vulnerability to enzymatic degradation and low immunogenicity of peptides. To further improve the potency of vaccine, we developed a poly(lactide-co-glycolide)-acid (PLGA) nanoparticle, which encapsulated the antigenic peptide HPV16 E744-62, and used adenosine triphosphate (ATP), one of the most important intracellular metabolites and an endogenous extracellular danger signal for the immune system, as a new adjuvant component. The results showed that PLGA nanoparticles increased the in vivo stability, lymph node accumulation, and dendritic cell (DC) uptake of the E7 peptide; in addition, ATP further increased the migration, nanoparticle uptake, and maturation of DCs. Preventive immunization with ATP-adjuvanted nanoparticles completely abolished the growth of TC-1 tumors in mice and produced long-lasting immunity against tumor rechallenge. When tumors were fully established, therapeutic immunization with ATP-adjuvanted nanoparticles still significantly inhibited tumor progression. Mechanistically, ATP-adjuvanted nanoparticles significantly improved the systemic generation of antitumor effector cells, boosted the local functional status of these cells in tumors, and suppressed the generation and tumor infiltration of immunosuppressive Treg cells and myeloid-derived suppressor cells. These findings indicate that ATP is an effective vaccine adjuvant and that nanoparticles adjuvanted with ATP were able to elicit robust antitumor cellular immunity, which may provide a promising therapeutic vaccine candidate for the treatment of clinical malignancies, such as cervical cancer.


Subject(s)
Adenosine Triphosphate/metabolism , Cancer Vaccines/immunology , Immunity, Cellular , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Neoplasms/therapy , Papillomavirus E7 Proteins/chemistry , Papillomavirus E7 Proteins/immunology , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Transplantation, Heterologous
5.
Acc Chem Res ; 53(10): 2094-2105, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33017150

ABSTRACT

The immune system has evolved over time to protect the host from foreign microorganisms. Activation of the immune system is predicated on a distinction between self and nonself. Unfortunately, cancer is characterized by genetic alterations in the host's cells, leading to uncontrolled cellular proliferation and evasion of immune surveillance. Cancer immunotherapy aims to educate the host's immune system to not only recognize but also attack and kill mutated cancer cells. While immune checkpoint blockers have been proven to be effective against multiple types of advanced cancer, the overall patient response rate still remains below 30%. Therefore, there is an urgent need to improve current cancer immunotherapies. In this Account, we present an overview of our recent progress on nanoparticle-based strategies for improving cancer vaccines and immunotherapies. We also present other complementary strategies to give a well-rounded snapshot of the field of combination cancer immunotherapy. The versatility and tunability of nanoparticles make them promising platforms for addressing individual challenges posed by various cancers. For example, nanoparticles can deliver cargo materials to specific cells, such as vaccines delivered to antigen-presenting cells for strong immune activation. Nanoparticles also allow for stimuli-responsive delivery of various therapeutics to cancer cells, thus forming the basis for combination cancer immunotherapy. Here, we focus on nanoparticle platforms engineered to deliver tumor antigens, whole tumor cells, and chemotherapeutic or phototherapeutic agents in a manner to effectively and safely trigger the host's immune system against tumor cells. For each work, we discuss the nanoparticle platform developed, synthesis chemistry, and in vivo applications. Nanovaccines offer a unique platform for codelivery of personalized tumor neoantigens and adjuvants and elicitation of robust immune responses against aggressive tumors. Nanovaccines either delivering whole tumor cell lysate or formed from tumor cell lysate may increase the repertoire of tumor antigens as immune targets while exploiting immunogenic cell death to prime antitumor immune responses. We also discuss how antigen- and whole tumor cell-based approaches may open the door for personalized cancer vaccination and immunotherapy. On the other hand, chemotherapy, phototherapy, and radiotherapy are more standardized cancer therapies, and nanoparticle-based approaches may promote their ability to initiate T cell activation against tumor cells and improve antitumor efficacy with minimal toxicity. Finally, building on the recent progress in nanoparticle-based cancer immunotherapy, the field should set the ultimate goal to be clinical translation and clinical efficacy. We will discuss regulatory, analytical, and manufacturing hurdles that should be addressed to expedite the clinical translation of nanomedicine-based cancer immunotherapy.


Subject(s)
Cancer Vaccines/administration & dosage , Immunotherapy , Nanoparticles/chemistry , Neoplasms/therapy , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Graphite/chemistry , Humans , Neoplasms/immunology , Neoplasms/prevention & control , Polymers/chemistry
6.
Pharmacol Ther ; 213: 107587, 2020 09.
Article in English | MEDLINE | ID: mdl-32470470

ABSTRACT

The widespread coronavirus SARS-CoV-2 has already infected over 4 million people worldwide, with a death toll over 280,000. Current treatment of COVID-19 patients relies mainly on antiviral drugs lopinavir/ritonavir, arbidol, and remdesivir, the anti-malarial drugs hydroxychloroquine and chloroquine, and traditional Chinese medicine. There are over 2,118 on-going clinical trials underway, but to date none of these drugs have consistently proven effective. Cathepsin L (CatL) is an endosomal cysteine protease. It mediates the cleavage of the S1 subunit of the coronavirus surface spike glycoprotein. This cleavage is necessary for coronavirus entry into human host cells, virus and host cell endosome membrane fusion, and viral RNA release for next round of replication. Here we summarize data regarding seven CatL-selective inhibitors that block coronavirus entry into cultured host cells and provide a mechanism to block SARS-CoV-2 infection in humans. Given the rapid growth of the SARS-CoV-2-positive population worldwide, ready-to-use CatL inhibitors should be explored as a treatment option. We identify ten US FDA-approved drugs that have CatL inhibitory activity. We provide evidence that supports the combined use of serine protease and CatL inhibitors as a possibly safer and more effective therapy than other available therapeutics to block coronavirus host cell entry and intracellular replication, without compromising the immune system.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cathepsin L/antagonists & inhibitors , Coronavirus Infections/drug therapy , Coronavirus Infections/physiopathology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/physiopathology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antigen-Presenting Cells/metabolism , Antimalarials/pharmacology , Antimalarials/therapeutic use , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Betacoronavirus , COVID-19 , Clinical Trials as Topic/statistics & numerical data , Dose-Response Relationship, Drug , Drug Approval , Drug Therapy, Combination , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Medicine, Chinese Traditional/methods , Pandemics , SARS-CoV-2 , Serine Endopeptidases/metabolism , United States , United States Food and Drug Administration
7.
J Immunotoxicol ; 17(1): 59-66, 2020 12.
Article in English | MEDLINE | ID: mdl-32091282

ABSTRACT

Injection site reaction (ISR) is a common side-effect associated with the use of peptide or protein pharmaceuticals. These types of pharmaceuticals-induced activation of antigen-presenting cells is assumed to be a key step in the pathogenesis of immune-mediated ISR. The present study was designed to evaluate the immunostimulatory properties of peptide or protein pharmaceuticals using human monocytic THP-1 cells. Here, THP-1 cells, with or without phorbol-12-myristate-13-acetate (PMA) pretreatment, were exposed to enfuvirtide and glatiramer acetate (positive controls) or evolocumab (negative control) for 6 or 24 h. PMA treatment differentiated non-adherent monocytic THP-1 (nTHP-1) cells into adherent macrophagic THP-1 (pTHP-1) cells that highly express CD11b and CD36. Enfuvirtide increased the release of cytokines, e.g. TNFα, MIP-1ß, and MCP-1, and expression of CD86 and CD54 on nTHP-1 cells at 24 h. Similar immunostimulatory properties of glatiramer acetate were observed both in the nTHP-1 and pTHP-1 cells at 6 h, but the responses were very weak in the pTHP-1 cells. Evolocumab did not affect cytokine secretion or cell surface marker expression in either cell type. Taken together, these in vitro THP-1 cell assays revealed the immunostimulatory properties of enfuvirtide and glatiramer acetate. This assay platform thus could serve as a powerful tool in evaluating potential immune-related ISR risks of peptide or protein pharmaceuticals in humans.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antigen-Presenting Cells/immunology , Enfuvirtide/immunology , Glatiramer Acetate/immunology , Injection Site Reaction/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Antigen Presentation/drug effects , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Cytokines/metabolism , Drug Evaluation, Preclinical/methods , Enfuvirtide/administration & dosage , Glatiramer Acetate/administration & dosage , Humans , Injections, Subcutaneous/adverse effects , THP-1 Cells
8.
Life Sci Alliance ; 2(1)2019 02.
Article in English | MEDLINE | ID: mdl-30665948

ABSTRACT

Chikungunya virus (CHIKV) has been a worldwide threat since its reemergence in La Reunion Island in 2004. Expression of the interferon-stimulated protein Viperin correlates with viral load burden in patients, and studies in mice have demonstrated its role to limit disease severity against CHIKV infection. Using Viperin -/- mice, we aimed to understand the contribution of Viperin to the T-cell immune response against CHIKV. CD4 T-cell depletion in Viperin -/- mice showed that increased late acute joint inflammation (5-8 d postinfection) was exclusively mediated by T cells. Specifically, CHIKV-infected Viperin -/- mice showed an increased INFγ Th1 profile of CD4 T cells, enhanced INFγ stimulation by APCs, an increased INFγ secretion profile in the joint microenvironment, and increased numbers of inflammatory monocytes in virus-infected joints compared with WT mice. Bone marrow grafting experiments showed that Viperin expression in both hematopoietic and non-hematopoietic cells is instrumental in reducing disease severity associated with a CD4 T-cell response.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Interferon-gamma/metabolism , Proteins/metabolism , Animals , Antigen-Presenting Cells/metabolism , Arthritis/metabolism , Arthritis/virology , Bone Marrow Transplantation , Chikungunya virus/isolation & purification , Female , Gene Knockout Techniques , HEK293 Cells , Humans , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Proteins/genetics , Viral Load
9.
Biomaterials ; 175: 82-92, 2018 08.
Article in English | MEDLINE | ID: mdl-29803106

ABSTRACT

Silica based nanoparticles have emerged as a promising vaccine delivery system for cancer immunotherapy, but their bio-degradability, adjuvanticity and the resultant antitumor activity remain to be largely improved. In this study, we report biodegradable glutathione-depletion dendritic mesoporous organosilica nanoparticles (GDMON) with a tetrasulfide-incorporated framework as a novel co-delivery platform in cancer immunotherapy. Functionalized GDMON are capable of co-delivering an antigen protein (ovalbumin) and a toll-like receptor 9 (TLR9) agonist into antigen presenting cells (APCs) and inducing endosome escape. Moreover, decreasing the intracellular glutathione (GSH) level through the -S-S-/GSH redox chemistry increases the ROS generation level both in vitro and in vivo, facilitating cytotoxic T lymphocyte (CTL) proliferation and reducing tumour growth in an aggressive B16-OVA melanoma tumour model. Our results have shown the potential of GDMON as a novel self-adjuvant and co-delivery nanocarrier for cancer vaccine.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/administration & dosage , Glutathione/metabolism , Melanoma, Experimental/therapy , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , Antigen-Presenting Cells/metabolism , Cell Line, Tumor , Cell Proliferation , Endosomes/physiology , Humans , Immunotherapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Oligodeoxyribonucleotides/administration & dosage , Ovalbumin/administration & dosage , Oxidation-Reduction , Polyethyleneimine/chemistry , Porosity , T-Lymphocytes, Cytotoxic/pathology , Toll-Like Receptor 9/agonists
10.
Innate Immun ; 24(4): 192-202, 2018 05.
Article in English | MEDLINE | ID: mdl-29631470

ABSTRACT

APCs such as monocytes and dendritic cells are among the first cells to recognize invading pathogens and initiate an immune response. The innate response can either eliminate the pathogen directly, or through presentation of Ags to T cells, which can help to clear the infection. Mucosal-associated invariant T (MAIT) cells are among the unconventional T cells whose activation does not involve the classical co-stimulation during Ag presentation. MAIT cells can be activated either via presentation of unconventional Ags (such as riboflavin metabolites) through the evolutionarily conserved major histocompatibility class I-like molecule, MR1, or directly by cytokines such as IL-12 and IL-18. Given that APCs produce cytokines and can express MR1, these cells can play an important role in both pathways of MAIT cell activation. In this review, we summarize evidence on the role of APCs in MAIT cell activation in infectious disease and cancer. A better understanding of the interactions between APCs and MAIT cells is important in further elucidating the role of MAIT cells in infectious diseases, which may facilitate the design of novel interventions such as vaccines.


Subject(s)
Antigen-Presenting Cells/immunology , Communicable Diseases/immunology , Lymphocyte Activation , Mucosal-Associated Invariant T Cells/immunology , Neoplasms/immunology , Antigen Presentation , Antigen-Presenting Cells/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Interleukin-12/metabolism , Interleukin-18/metabolism , Mucosal-Associated Invariant T Cells/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism
11.
Parasite ; 25: 11, 2018.
Article in English | MEDLINE | ID: mdl-29533762

ABSTRACT

Tectona grandis (teak) and Vernonia amygdalina (bitter leaf) are plants used in traditional medicine in West Africa. In this study, we tested ethanolic and hydro-ethanolic extracts of bark and leaves of T. grandis and ethanolic extract of leaves of V. amygdalina for their inhibitory effect on Toxoplasma gondii, a protozoan parasite responsible for toxoplasmosis. Ethanolic extract of V. amygdalina leaves had proportional contents of phenols, tannins, flavonoids, and polysaccharides. This extract presented the highest efficacy against T. gondii, the lowest cytotoxicity to mammalian cells, but moderate anti-oxidant activity compared to other plant extracts. Ethanolic extract of T. grandis bark also had elevated anti-T. gondii activity, low cytotoxicity on mammalian cells, and one of the highest anti-oxidant activities. However, the phytochemical content of this extract was not very different from the hydro-ethanolic extract, which had no anti-T. gondii activity. In addition, ethanolic extract of V. amygdalina leaves, but not of T. grandis bark, significantly increased the production of TNF-α and NO by antigen-presenting cells. Both extracts had the tendency to decrease expression of major histocompatibility complex molecules at the surface of antigen-presenting cells, while they did not modulate the percentage of apoptotic cells. A study of signalling pathways would help to determine the mechanisms of action of these plant extracts.


TITLE: Les extraits de Tectona grandis et de Vernonia amygdalina ont des propriétés anti-Toxoplasma et pro-inflammatoires in vitro. ABSTRACT: Tectona grandis (teck) et Vernonia amygdalina sont des plantes utilisées dans la médecine traditionnelle en Afrique de l'Ouest. Dans cette étude, l'effet inhibiteur d'extraits éthanoliques et hydro-éthanoliques d'écorce et de feuilles de T. grandis et de l'extrait éthanolique des feuilles de V. amygdalina a été étudié sur Toxoplasma gondii, un parasite protozoaire responsable de la toxoplasmose. L'extrait éthanolique des feuilles de V. amygdalina avait des quantités équivalentes de phénols, tanins, flavonoïdes et polysaccharides. Cet extrait présentait la plus grande efficacité contre T. gondii, la plus faible cytotoxicité vis-à-vis de cellules de mammifères, mais une activité anti-oxydante moyenne comparée aux autres extraits de plantes. L'extrait éthanolique d'écorce de T. grandis avait aussi une activité anti-T. gondii élevée, une faible cytotoxicité vis-à-vis des cellules de mammifères et l'une des activités anti-oxydantes les plus élevées. Cependant, le contenu phytochimique de cet extrait n'était pas très différent de l'extrait hydro-éthanolique qui n'avait pas d'activité anti-T. gondii. De plus, l'extrait éthanolique des feuilles de V. amygdalina, mais pas de l'écorce de T. grandis, augmentait significativement la production de TNF-α et de NO par les cellules présentatrices d'antigènes. Les deux extraits avaient tendance à diminuer l'expression des molécules du complexe majeur d'histocompatibilité à la surface des cellules présentatrices d'antigènes alors qu'ils ne modulaient pas le pourcentage de cellules apoptotiques. L'étude des voies de signalisation permettrait de comprendre les mécanismes d'action de ces extraits de plantes.


Subject(s)
Lamiaceae/chemistry , Plant Extracts/pharmacology , Toxoplasma/drug effects , Vernonia/chemistry , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Antioxidants/metabolism , Apoptosis/drug effects , Major Histocompatibility Complex/drug effects , Mammals , Nitric Oxide/metabolism , Plant Bark/chemistry , Plant Extracts/toxicity , Plant Leaves/chemistry , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
12.
Adv Healthc Mater ; 6(17)2017 Sep.
Article in English | MEDLINE | ID: mdl-28557331

ABSTRACT

Mesoporous silica nanoparticles are reported as adjuvants in nanovaccines in generating robust antigen-specific immunity. However, the effect of surface chemistry in initiating and modulating the immune response remains largely unexplored. In this study, mesoporous silica nanorods (MSNRs) are modified with NH2 and C18 groups to investigate the influence of surface functional groups (OH, NH2 , and C18 ) on their adjuvant efficacy. It is found that compared to OH and NH2 groups, the hydrophobic C18 modification significantly enhances antigen uptake by antigen presenting cells and endosomal-lysosomal escape in vitro, dendritic cells, and macrophages maturation ex vivo, and elicits secretion of interferon-γ level and antibody response in immunized mice. Moreover, bare MSNR and MSNRNH2 exhibit T-helper 2 biased immune response, while MSNRC18 shows a T-helper 1 biased immune response. These findings suggest that the surface chemistry of nanostructured adjuvants has profound impact on the immune response, which provides useful guidance for the design of effective nanomaterial based vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Nanotubes/chemistry , Silicon Dioxide/chemistry , Animals , Antibody Formation/drug effects , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Antigens/metabolism , Cell Death/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endocytosis/drug effects , Immunization , Immunoglobulin G/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Nanotubes/ultrastructure , Ovalbumin/metabolism , Porosity , RAW 264.7 Cells , Spleen/cytology , Surface Properties , Vaccines
13.
Methods Mol Biol ; 1371: 43-78, 2016.
Article in English | MEDLINE | ID: mdl-26530794

ABSTRACT

FOXP3+ T-regulatory (Treg) cells have important roles in immune homeostasis, and alterations in their number and function can predispose to diseases ranging from autoimmunity to allograft rejection and tumor growth. Reliable identification of human Tregs remains a persistent problem due to a lack of specific markers. The most definitive Treg characterization currently involves combined assessment of phenotypic, epigenetic and functional parameters, with the latter typically involving in vitro Treg suppression assays. Unfortunately, suppression assays are frequently performed using differing methods and readouts, limiting comparisons between studies. We provide a perspective on our experience with human and murine Treg suppression assay conditions, including Treg data obtained in clinical transplant studies, Tregs isolated from healthy donors and treated with epigenetically active compounds, and Tregs from standard murine strains (C57BL/6 and BALB/c). We provide detailed descriptions and illustrations of typical problems, shortcomings and troubleshooting; describe new modifications and approaches; and present a new method for calculation of suppressive assay data using a modified area-under-curve (AUC) method. This method allows us to directly compare Treg suppressive function between multiple patients (such as in clinical transplant studies), to reliably track changes in Treg function from the same person over time, or compare effects of Treg-modulating compounds tested with different healthy donors Tregs in separate or combined experimental settings.


Subject(s)
Flow Cytometry/methods , Flow Cytometry/standards , Immunomodulation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Surface/metabolism , Area Under Curve , Cell Separation/methods , Cryopreservation , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Humans , Immunomodulation/drug effects , Immunophenotyping , Mice , Phenotype , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects
14.
Allergy ; 70(4): 408-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25631199

ABSTRACT

BACKGROUND: A detailed characterization of human oral immune cells is needed to better understand local mechanisms associated with allergen capture following oral exposure. METHODS: Oral immune cells were characterized by immunohistology and immunofluorescence in biopsies obtained from three healthy individuals and 23 birch pollen-allergic patients with/without oral allergy syndrome (OAS), at baseline and after 5 months of sublingual allergen immunotherapy (AIT). RESULTS: Similar cell subsets (i.e., dendritic cells, mast cells, and T lymphocytes) were detected in oral tissues from healthy and birch pollen-allergic individuals. CD207+ Langerhans cells (LCs) and CD11c+ myeloid dendritic cells (DCs) were found in both the epithelium and the papillary layer of the Lamina propria (LP), whereas CD68+ macrophages, CD117+ mast cells, and CD4+ /CD8+ T cells were rather located in both the papillary and reticular layers of the LP. Patterns of oral immune cells were identical in patients with/without OAS, except lower numbers of CD207+ LCs found in oral tissues from patients with OAS, when compared to OAS- patients (P < 0.05). A 5-month sublingual AIT had a limited impact on oral immune cells, with only a significant increase in IgE+ cells in patients from the active group. Colocalization experiments confirmed that such IgE-expressing cells mostly encompass CD68+ macrophages located in the LP, and to a lesser extent CD207+ LCs in the epithelium. CONCLUSION: Two cell subsets contribute to antigen/allergen uptake in human oral tissues, including (i) CD207+ LCs possibly involved in the physiopathology of OAS and (ii) CD68+ macrophages likely critical in allergen capture via IgE-facilitated mechanisms during sublingual AIT.


Subject(s)
Allergens/immunology , Antigen-Presenting Cells/immunology , Betula , Pollen/immunology , Rhinitis, Allergic, Seasonal/immunology , Antigen-Presenting Cells/metabolism , Antigens, Surface/metabolism , Biomarkers , Biopsy , Case-Control Studies , Female , Gene Expression , Gingiva/immunology , Gingiva/metabolism , Gingiva/pathology , Humans , Hypersensitivity/diagnosis , Hypersensitivity/immunology , Hypersensitivity/therapy , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunophenotyping , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/immunology , Mast Cells/metabolism , Rhinitis, Allergic, Seasonal/diagnosis , Rhinitis, Allergic, Seasonal/therapy , Sublingual Immunotherapy , Syndrome , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
15.
Food Chem ; 164: 36-43, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24996302

ABSTRACT

Polyphenols, the potent plant secondary metabolites, have beneficial effects on human health, but the mechanism(s) by which these effects are exerted is not well understood. Here, we present the detailed analysis of the interactions between the major green tea catechin, epigallo-catechin 3-gallate (EGCG), and the major dietary protein and allergen, ovalbumin (OVA). We show that EGCG binds to the pocket that partly overlaps with the previously identified IgE-binding region in OVA, and that this interaction induces structural changes in the allergen. Moreover, our ex vivo studies reveal that OVA binds IgE and stimulates degranulation of basophils, and that its uptake by monocytes proceeds at a slower rate in the presence of EGCG. This study provides further evidence in support of the proposed mechanism by which EGCG interactions with the food allergens contribute to its diverse biological activities and may impair antigen uptake by antigen-presenting cells.


Subject(s)
Catechin/analogs & derivatives , Egg White/chemistry , Ovalbumin/chemistry , Allergens/chemistry , Antigen-Presenting Cells/metabolism , Basophils/metabolism , Catechin/chemistry , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Food Hypersensitivity , Humans , Immunoglobulin E/chemistry , Monocytes/metabolism , Polyphenols , Protein Binding , Protein Conformation , Spectrometry, Fluorescence , Tea/chemistry
16.
J Immunol ; 192(7): 3259-68, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24610009

ABSTRACT

Adjuvants are an essential component of modern vaccines and used for their ability to elicit immunity to coadministered Ags. Many adjuvants in clinical development are particulates, but how they drive innate and adaptive immune responses remains poorly understood. Studies have shown that a number of vaccine adjuvants activate inflammasome pathways in isolated APCs. However, the contribution of inflammasome activation to vaccine-mediated immunity in vivo remains controversial. In this study, we evaluated immune cell responses to the ISCOMATRIX adjuvant (IMX) in mice. Like other particulate vaccine adjuvants, IMX potently activated the NALP-3-ASC-Caspase-1 inflammasome in APCs, leading to IL-1ß and IL-18 production. The IL-18R pathway, but not IL-1R, was required for early innate and subsequent cellular immune responses to a model IMX vaccine. APCs directly exposed to IMX underwent an endosome-mediated cell-death response, which we propose initiates inflammatory events locally at the injection site. Importantly, both inflammasome-related and -unrelated pathways contributed to IL-18 dependence in vivo following IMX administration. TNF-α provided a physiological priming signal for inflammasome-dependent IL-18 production by APCs, which correlated with reduced vaccine-mediated immune cell responses in TNF-α- or TNFR-deficient mice. Taken together, our findings highlight an important disconnect between the mechanisms of vaccine adjuvant action in vitro versus in vivo.


Subject(s)
Cholesterol/immunology , Immunity/immunology , Inflammasomes/immunology , Interleukin-18/immunology , Phospholipids/immunology , Saponins/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Blotting, Western , Cell Survival/drug effects , Cell Survival/immunology , Cholesterol/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Combinations , Humans , Immunity/drug effects , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lysosomes/drug effects , Lysosomes/immunology , Lysosomes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phospholipids/pharmacology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Saponins/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
17.
Expert Opin Ther Targets ; 18(5): 513-25, 2014 May.
Article in English | MEDLINE | ID: mdl-24568095

ABSTRACT

INTRODUCTION: Psoriasis is a chronic inflammatory skin disorder determined by the activation of several immune cells and resident tissue cells. Various cytokines mediate inflammatory signals, including IL-23, which is an important factor involved in the differentiation of T helper (Th17) cells. AREAS COVERED: Increasing evidence suggests that IL-23 is a central cytokine to the pathogenesis of psoriasis. An overview on both experimental and human data will be reported in order to support the hypothesis of a key pathogenic role of IL-23/Th17 axis. EXPERT OPINION: Targeting IL-23 might be a more selective, valid and effective therapeutic approach, which, potentially, may show important advantages in terms of long-term efficacy and safety in the treatment of psoriasis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interleukin-23/physiology , Molecular Targeted Therapy , Psoriasis/physiopathology , Animals , Antigen-Presenting Cells/metabolism , Bacterial Infections/immunology , Cell Differentiation , Clinical Trials as Topic , Cytokines/physiology , Drug Evaluation, Preclinical , Genetic Predisposition to Disease , Humans , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-12 Subunit p40/genetics , Interleukin-23/antagonists & inhibitors , Interleukin-23/genetics , Interleukin-23 Subunit p19/antagonists & inhibitors , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/physiology , Keratinocytes/metabolism , Mice , Mice, Knockout , Psoriasis/genetics , Psoriasis/immunology , Receptors, Interleukin/antagonists & inhibitors , Receptors, Interleukin/physiology , Signal Transduction , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology
18.
PLoS One ; 8(10): e76716, 2013.
Article in English | MEDLINE | ID: mdl-24116141

ABSTRACT

The bark of Ulmus davidiana var. japonica Nakai (Ulmaceae) has been used in traditional Korean medicine for chronic inflammation in the gastrointestinal tract. Here we investigated the frequency and cytokine profile of the major immune cells in the small intestinal lamina propria (SI LP), spleen, and mesenteric lymph nodes (MLNs) of mice treated orally with Ulmus davidiana var. japonica Nakai bark water extract (UDE) to address the immunomodulatory role of this herb in intestinal homeostasis. B6 mice were given 5g/kg UDE once daily for 14 days. They were then sacrificed, and cells were isolated from the spleen, MLNs, and SI LP. The proportion of B versus T lymphocytes, CD4(+) versus CD8(+) T lymphocytes, Th1 and Th17 cells, and Foxp3(+) regulatory T cells in the spleen, MLNs, and SI LP were analyzed. The frequency of antigen-presenting cells (APCs), including dendritic cells, macrophages, and eosinophils in the SI LP and the expression of costimulatory molecules on APCs were also evaluated. The numbers and frequencies of Th1 and Th17 cells in the SI LP were significantly reduced in the UDE-treated mice compared with PBS controls. In addition, the proportion of IL-4-producing eosinophils in the SI LP was significantly elevated in the UDE-treated mice compared with controls. Taken together, these data indicate that UDE up-regulates the number and frequency of SI LP eosinophils, which can down-regulate the Th1 and Th17 responses via IL-4 secretion and contribute to intestinal homeostasis.


Subject(s)
Eosinophils/drug effects , Intestine, Small/drug effects , Plant Extracts/pharmacology , Th1 Cells/drug effects , Th17 Cells/drug effects , Ulmus/chemistry , Administration, Oral , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Count , Enzyme-Linked Immunosorbent Assay , Eosinophils/immunology , Eosinophils/metabolism , Female , Homeostasis/drug effects , Homeostasis/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interleukin-4/immunology , Interleukin-4/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plant Bark/chemistry , Plant Extracts/administration & dosage , Plant Extracts/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Up-Regulation/drug effects
19.
Nanoscale ; 5(12): 5167-79, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23657437

ABSTRACT

Vaccines have been at the forefront of improving human health for over two centuries. The challenges faced in developing effective vaccines flow from complexities associated with the immune system and requirement of an efficient and safe adjuvant to induce a strong adaptive immune response. Development of an efficient vaccine formulation requires careful selection of a potent antigen, efficient adjuvant and route of delivery. Adjuvants are immunological agents that activate the antigen presenting cells (APCs) and elicit a strong immune response. In the past decade, the use of mesoporous silica nanoparticles (MSNs) has gained significant attention as potential delivery vehicles for various biomolecules. In this review, we aim to highlight the potential of MSNs as vaccine delivery vehicles and their ability to act as adjuvants. We have provided an overview on the latest progress on synthesis, adsorption and release kinetics and biocompatibility of MSNs as next generation antigen carriers and adjuvants. A comprehensive summary on the ability of MSNs to deliver antigens and elicit both humoral and cellular immune responses is provided. Finally, we give insight on fundamental challenges and some future prospects of these nanoparticles as adjuvants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/administration & dosage , Drug Carriers/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , Antigen-Presenting Cells/metabolism , Cell Survival/drug effects , Humans , Nanoparticles/toxicity , Porosity
20.
PLoS One ; 8(3): e60068, 2013.
Article in English | MEDLINE | ID: mdl-23533665

ABSTRACT

BACKGROUND: To induce potent epitope-specific T cell immunity by a peptide-based vaccine, epitope peptides must be delivered efficiently to antigen-presenting cells (APCs) in vivo. Therefore, selecting an appropriate peptide carrier is crucial for the development of an effective peptide vaccine. In this study, we explored new peptide carriers which show enhancement in cytotoxic T lymphocyte (CTL) induction capability. METHODOLOGY/PRINCIPAL FINDINGS: Data from an epitope-specific in vivo CTL assay revealed that phosphatidylserine (PS) has a potent adjuvant effect among candidate materials tested. Further analyses showed that PS-conjugated antigens were preferentially and efficiently captured by professional APCs, in particular, by CD11c(+)CD11b(+)MHCII(+) conventional dendritic cells (cDCs) compared to multilamellar liposome-conjugates or unconjugated antigens. In addition, PS demonstrated the stimulatory capacity of peptide-specific helper T cells in vivo. CONCLUSIONS/SIGNIFICANCE: This work indicates that PS is the easily preparable efficient carrier with a simple structure that delivers antigen to professional APCs effectively and induce both helper and cytotoxic T cell responses in vivo. Therefore, PS is a promising novel adjuvant for T cell-inducing peptide vaccines.


Subject(s)
Antigen-Presenting Cells/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes/immunology , Phosphatidylserines/chemistry , Animals , Antigen-Presenting Cells/immunology , CD11b Antigen/metabolism , CD11c Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/immunology , Mice , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL