Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 996
Filter
1.
Molecules ; 29(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38675621

ABSTRACT

Allogeneic hematopoietic cell transplantation (allo-HCT) is a highly effective, well-established treatment for patients with various hematologic malignancies and non-malignant diseases. The therapeutic benefits of allo-HCT are mediated by alloreactive T cells in donor grafts. However, there is a significant risk of graft-versus-host disease (GvHD), in which the donor T cells recognize recipient cells as foreign and attack healthy organs in addition to malignancies. We previously demonstrated that targeting JAK1/JAK2, mediators of interferon-gamma receptor (IFNGR) and IL-6 receptor signaling, in donor T cells using baricitinib and ruxolitinib results in a significant reduction in GvHD after allo-HCT. Furthermore, we showed that balanced inhibition of JAK1/JAK2 while sparing JAK3 is important for the optimal prevention of GvHD. Thus, we have generated novel JAK1/JAK2 inhibitors, termed WU derivatives, by modifying baricitinib. Our results show that WU derivatives have the potential to mitigate GvHD by upregulating regulatory T cells and immune reconstitution while reducing the frequencies of antigen-presenting cells (APCs) and CD80 expression on these APCs in our preclinical mouse model of allo-HCT. In addition, WU derivatives effectively downregulated CXCR3 and T-bet in primary murine T cells. In summary, we have generated novel JAK inhibitors that could serve as alternatives to baricitinib or ruxolitinib.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Pyrazoles , Transplantation, Homologous , Animals , Mice , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Azetidines/pharmacology , Disease Models, Animal , Graft vs Host Disease/prevention & control , Graft vs Host Disease/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Mice, Inbred C57BL , Purines/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects
2.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163272

ABSTRACT

Polypod-like structured nucleic acids (polypodnas), which are nanostructured DNAs, are useful for delivering cytosine-phosphate guanine oligodeoxynucleotides (CpG ODNs) to antigen-presenting cells (APCs) expressing Toll-like receptor 9 (TLR9) for immune stimulation. Lipid modification is another approach to deliver ODNs to lymph nodes, where TLR9-positive APCs are abundant, by binding to serum albumin. The combination of these two methods can be useful for delivering CpG ODNs to lymph nodes in vivo. In the present study, CpG1668, a phosphodiester-type CpG ODN, was modified with stearic acid (SA) to obtain SA-CpG1668. Tripodna, a polypodna with three pods, was selected as the nanostructured DNA. Tripodnas loaded with CpG1668 or SA-CpG1668 were obtained in high yields. SA-CpG1668/tripodna bound more efficiently to plasma proteins than CpG1668/tripodna and was more efficiently taken up by macrophage-like RAW264.7 cells than CpG1668/tripodna, whereas the levels of tumor necrosis factor-α released from the cells were comparable between the two. After subcutaneous injection into mice, SA-CpG1668/tripodna induced significantly higher interleukin (IL)-12 p40 production in the draining lymph nodes than SA-CpG1668 or CpG1668/tripodna, with reduced IL-6 levels in plasma. These results indicate that the combination of SA modification and nanostructurization is a useful approach for the targeted delivery of CpG ODNs to lymph nodes.


Subject(s)
Antigen-Presenting Cells/metabolism , Nanostructures/chemistry , Oligodeoxyribonucleotides/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/drug effects , DNA/immunology , Drug Delivery Systems/methods , Female , Immunization/methods , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Nanostructures/therapeutic use , Nucleic Acid Conformation/drug effects , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/metabolism , Proof of Concept Study , RAW 264.7 Cells , Stearic Acids/chemistry
3.
J Mater Chem B ; 9(37): 7713-7724, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34545900

ABSTRACT

Induction of cellular immunity is important for effective cancer immunotherapy. Although various antigen carriers for cancer immunotherapy have been developed to date, balancing efficient antigen delivery to antigen presenting cells (APCs) and their activation via innate immune receptors, both of which are crucially important for the induction of strong cellular immunity, remains challenging. For this study, branched ß-glucan was selected as an intrinsically immunity-stimulating and biocompatible material. It was engineered to develop multifunctional liposomal cancer vaccines capable of efficient interactions with APCs and subsequent activation of the cells. Hydroxy groups of branched ß-glucan (Aquaß) were modified with 3-methylglutaric acid ester and decyl groups, respectively, to provide pH-sensitivity and anchoring capability to the liposomal membrane. The modification efficiency of Aquaß derivatives to the liposomes was significantly high compared with linear ß-glucan (curdlan) derivatives. Aquaß derivative-modified liposomes released their contents in response to weakly acidic pH. As a model antigenic protein, ovalbumin (OVA)-loaded liposomes modified with Aquaß derivatives interacted efficiently with dendritic cells, and induced inflammatory cytokine secretion from the cells. Subcutaneous administration of Aquaß derivative-modified liposomes suppressed the growth of the E.G7-OVA tumor significantly compared with curdlan derivative-modified liposomes. Aquaß derivative-modified liposomes induced the increase of CD8+ T cells, and polarized macrophages to the antitumor M1-phenotype within the tumor microenvironment. Therefore, pH-sensitive Aquaß derivatives can be promising materials for liposomal antigen delivery systems to induce antitumor immune responses efficiently.


Subject(s)
Antigen-Presenting Cells/immunology , Biocompatible Materials/chemistry , Liposomes/chemistry , beta-Glucans/chemistry , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Biocompatible Materials/pharmacology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/metabolism , Female , Hydrogen-Ion Concentration , Immunity, Cellular , Immunotherapy , Macrophage Activation , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Neoplasms/therapy , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Tumor Microenvironment , beta-Glucans/metabolism
4.
Nanomedicine ; 37: 102442, 2021 10.
Article in English | MEDLINE | ID: mdl-34284132

ABSTRACT

Contractile forces within the planar interface between T cell and antigen-presenting surface mechanically stimulate T cell receptors (TCR) in the mature immune synapses. However, the origin of mechanical stimulation during the initial, i.e., presynaptic, microvilli-based TCR activation in the course of immune surveillance remains unknown and new tools to help address this problem are needed. In this work, we develop nucleic acid nanoassembly (NAN)-based technology for functionalization of hydrogels using isothermal toehold-mediated reassociation of RNA/DNA heteroduplexes. Resulting platform allows for regulation with NAN linkers of 3D force momentum along the TCR mechanical axis, whereas hydrogels contribute to modulation of 2D shear modulus. By utilizing different lengths of NAN linkers conjugated to polyacrylamide gels of different shear moduli, we demonstrate an efficient capture of human T lymphocytes and tunable activation of TCR, as confirmed by T-cell spreading and pY foci.


Subject(s)
Hydrogels/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Receptors, Antigen, T-Cell/genetics , Antigen-Presenting Cells/drug effects , DNA/chemistry , DNA/pharmacology , Humans , Hydrogels/chemistry , Lymphocyte Activation/genetics , Lymphocytes/metabolism , Nucleic Acid Heteroduplexes/chemistry , Nucleic Acid Heteroduplexes/genetics , Nucleic Acid Heteroduplexes/pharmacology , RNA/chemistry , RNA/genetics , Receptors, Antigen, T-Cell/drug effects , T-Lymphocytes/drug effects
5.
Sci Rep ; 11(1): 13912, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34230534

ABSTRACT

Anti-PD-L1 antibodies benefit many cancer patients, even those with "non-inflamed tumor". Determining which patients will benefit remains an important clinical goal. In a non-inflamed tumor mouse model, we found that PD-L1 was highly expressed on antigen-presenting cells (APCs) especially on CD103+ CD11c+ dendritic cells in tumor-draining lymph nodes (dLNs), suppressing T-cell priming by APCs. In this model, anti-PD-L1 antibodies enhanced T-cell priming and increased CXCR3+ activated T-cells in dLNs, which was followed by the trafficking of T-cells to tumors in response to CXCR3 ligands. As predictive biomarker, each APCs-related gene expression (AP score) alone or T-cells trafficking-related chemokine gene expression (T score) alone were still less than perfect among the 17 mouse models examined. However a combining score of AP score and T score (AP/T score) precisely identified anti-PD-L1-sensitive tumors. In the phase 3 trial of atezolizumab vs docetaxel in advanced NSCLC patients (OAK), the AP/T score could identify atezolizumab-treated NSCLC patients who achieved significant improvement in overall survival. This biomarker concept would be a clinically valuable for prediction of anti-PD-L1 antibody efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cell Movement , Cross-Priming/immunology , Lymph Nodes/immunology , Receptors, CXCR3/metabolism , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Line, Tumor , Cell Movement/drug effects , Cross-Priming/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Gene Expression Regulation, Neoplastic/drug effects , Ligands , Lymph Nodes/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Models, Biological , T-Lymphocytes/drug effects
6.
Adv Drug Deliv Rev ; 179: 113905, 2021 12.
Article in English | MEDLINE | ID: mdl-34331988

ABSTRACT

Immunotherapy that utilizes the human immune system to fight cancer represents a revolutionary method for cancer treatment. Immunotherapeutic agents that trigger the immune response should be carefully delivered to the desired site to maximize immunotherapy effectiveness and minimize side effects. Vesicles offer the possibility of encapsulating both hydrophilic and hydrophobic drugs and thus serve as a promising delivery tool. As multiple irreconcilable requirements exist at different transport stages, developing vesicles transformable in response to given stimuli is of great significance. In this review, we first introduced various vesicle types used for immunotherapy. Furthermore, the typical stimuli that trigger vesicle transformation and the usually generated transformation styles were described. Focusing on three aspects of antigen-presenting cell (APC)/T cell activation, tumor microenvironment (TME) amelioration, and immunogenic cell death (ICD)-induced immunotherapy, we reviewed recently reported transformable vesicles for tumor treatment. Finally, we put forward possible directions for future research and clinical translation.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Drug Delivery Systems/methods , Immunogenic Cell Death/drug effects , Tumor Microenvironment/drug effects , Antigen-Presenting Cells/drug effects , Chemistry, Pharmaceutical , Humans , T-Lymphocytes/drug effects
7.
Front Immunol ; 12: 645962, 2021.
Article in English | MEDLINE | ID: mdl-34122406

ABSTRACT

The failure of M. bovis BCG to induce long-term protection has been endowed to its inability to escape the phagolysosome, leading to mild activation of CD8+ mediated T cell response. Induction of apoptosis in host cells plays an important role in potentiating dendritic cells-mediated priming of CD8+ T cells, a process defined as "cross-priming." Moreover, IL-10 secretion by infected cells has been reported to hamper BCG-induced immunity against Tuberculosis (TB). Previously, we have reported that apoptosis of BCG-infected macrophages and inhibition of IL-10 secretion is FOXO3 dependent, a transcription factor negatively regulated by the pro-survival activated threonine kinase, Akt. We speculate that FOXO3-mediated induction of apoptosis and abrogation of IL-10 secretion along with M. bovis BCG immunization might enhance the protection imparted by BCG. Here, we have assessed whether co-administration of a known anti-cancer Akt inhibitor, MK-2206, enhances the protective efficacy of M. bovis BCG in mice model of infection. We observed that in vitro MK-2206 treatment resulted in FOXO3 activation, enhanced BCG-induced apoptosis of macrophages and inhibition of IL-10 secretion. Co-administration of M. bovis BCG along with MK-2206 also increased apoptosis of antigen-presenting cells in draining lymph nodes of immunized mice. Further, MK-2206 administration improved BCG-induced CD4+ and CD8+ effector T cells responses and its ability to induce both effector and central memory T cells. Finally, we show that co-administration of MK-2206 enhanced the protection imparted by M. bovis BCG against Mtb in aerosol infected mice and guinea pigs. Taken together, we provide evidence that MK-2206-mediated activation of FOXO3 potentiates BCG-induced immunity and imparts protection against Mtb through enhanced innate immune response.


Subject(s)
BCG Vaccine/immunology , Heterocyclic Compounds, 3-Ring/pharmacology , Tuberculosis/prevention & control , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/physiology , Apoptosis/drug effects , Cells, Cultured , Forkhead Box Protein O3/physiology , Guinea Pigs , Immunologic Memory/drug effects , Macrophages/microbiology , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
8.
Front Immunol ; 12: 667478, 2021.
Article in English | MEDLINE | ID: mdl-34025669

ABSTRACT

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antigen-Presenting Cells/drug effects , Brain/drug effects , Encephalitis, Viral/prevention & control , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Macrophages/drug effects , Rhabdoviridae Infections/prevention & control , Vesiculovirus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Brain/immunology , Brain/metabolism , Brain/virology , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/metabolism , Encephalitis, Viral/virology , HEK293 Cells , Hepatitis A Virus Cellular Receptor 2/immunology , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions , Humans , Injections, Intraperitoneal , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RAW 264.7 Cells , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/metabolism , Rhabdoviridae Infections/virology , Ubiquitination , Vero Cells , Vesiculovirus/pathogenicity , Viral Load
9.
Front Immunol ; 12: 649135, 2021.
Article in English | MEDLINE | ID: mdl-33841433

ABSTRACT

The perfect synchronization of maternal immune-endocrine mechanisms and those of the fetus is necessary for a successful pregnancy. In this report, decidual immune cells at the maternal-fetal interface were detected that expressed TIGIT (T cell immunoreceptor with Ig and ITIM domains), which is a co-inhibitory receptor that triggers immunological tolerance. We generated recombinant TIGIT-Fc fusion proteins by linking the extracellular domain of TIGIT and silent Fc fragments. The treatment with TIGIT-Fc of human decidual antigen presenting cells (APCs), the decidual dendritic cells (dDCs), and decidual macrophages (dMϕs) increased the production of interleukin 10 and induced the decidua APCs to powerfully polarize the decidual CD4+ T cells toward a classic TH2 phenotype. We further proposed that Notch signaling shows a pivotal effect on the transcriptional regulation in decidual immune cell subsets. Moreover, the administration of TIGIT-Fc to CBA/J pregnant mice at preimplantation induced CD4+ forkhead box P3+ (Foxp3+) regulatory T cells and tolerogenic dendritic cells and increased pregnancy rates in an abortion-prone animal model stress. The results suggested the therapeutic potential of the TIGIT-Fc fusion protein in reinstating immune tolerance in failing pregnancies.


Subject(s)
Decidua/immunology , Immune Tolerance/immunology , Immunoglobulin Fc Fragments/immunology , Maternal-Fetal Exchange/immunology , Receptors, Immunologic/immunology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Decidua/cytology , Decidua/drug effects , Decidua/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Humans , Immune Tolerance/drug effects , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/therapeutic use , Interleukin-10/immunology , Interleukin-10/metabolism , Lymphocyte Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Maternal-Fetal Exchange/drug effects , Mice, Inbred CBA , Mice, Inbred DBA , Pregnancy , Receptors, Immunologic/chemistry , Receptors, Immunologic/therapeutic use
10.
Mol Nutr Food Res ; 65(12): e2001034, 2021 06.
Article in English | MEDLINE | ID: mdl-33890400

ABSTRACT

INTRODUCTION: Lunasin is a soybean bioactive peptide with a variety of beneficial properties against chronic disorders. However, its effect in human primary intestinal cells remains unknown. Hence, this study aims to characterize its ex vivo biological activity in the human intestinal mucosa. METHODS AND RESULTS: Human intestinal biopsies, obtained from healthy controls, are ex vivo conditioned with lunasin both in the presence/absence of lipopolysaccharide (LPS). Peptide maintains its stability during biopsy culture by HPLC-MS/MS analysis. Lunasin is bioactive in the human mucosa, as it induces IL-1ß, TNF-α, IL-17A, CCL2, and PGE2/COX-2 gene expression together with an increased expression of tolerogenic IL-10 and TGFß, while it also downregulates the expression of iNOS and subunit p65 from NF-κB. Indeed, lunasin also abrogates the LPS-induced pro-inflammatory response, downregulating IL-17A, IFNγ, and IL-8 expression, and inducing IL-10 and TGFß expression. These results are also mirrored in the cell-free culture supernatants at the protein level by Multiplex. Moreover, lunasin further induces a regulatory phenotype and function on human intestinal conventional dendritic cell and macrophage subsets as assessed by flow cytometry. CONCLUSIONS: We hereby have characterized lunasin as an immunomodulatory peptide with potential capacity to prevent immune and inflammatory-mediated disorders in the human gastrointestinal tract.


Subject(s)
Gene Expression Regulation/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Soybean Proteins/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antigen-Presenting Cells/drug effects , Cytokines/metabolism , Humans , Lipopolysaccharides/pharmacology , Soybean Proteins/immunology
11.
Sci Rep ; 11(1): 5402, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33686135

ABSTRACT

Most multiple sclerosis (MS) patients given currently available disease-modifying drugs (DMDs) experience progressive disability. Accordingly, there is a need for new treatments that can limit the generation of new waves T cell autoreactivity that drive disease progression. Notably, immune cells express GABAA-receptors (GABAA-Rs) whose activation has anti-inflammatory effects such that GABA administration can ameliorate disease in models of type 1 diabetes, rheumatoid arthritis, and COVID-19. Here, we show that oral GABA, which cannot cross the blood-brain barrier (BBB), does not affect the course of murine experimental autoimmune encephalomyelitis (EAE). In contrast, oral administration of the BBB-permeable GABAA-R-specific agonist homotaurine ameliorates monophasic EAE, as well as advanced-stage relapsing-remitting EAE (RR-EAE). Homotaurine treatment beginning after the first peak of paralysis reduced the spreading of Th17 and Th1 responses from the priming immunogen to a new myelin T cell epitope within the CNS. Antigen-presenting cells (APC) isolated from homotaurine-treated mice displayed an attenuated ability to promote autoantigen-specific T cell proliferation. The ability of homotaurine treatment to limit epitope spreading within the CNS, along with its safety record, makes it an excellent candidate to help treat MS and other inflammatory disorders of the CNS.


Subject(s)
Central Nervous System/pathology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Taurine/analogs & derivatives , Animals , Antigen Presentation/drug effects , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Cell Proliferation/drug effects , Central Nervous System/drug effects , Central Nervous System/immunology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Recurrence , Spleen/pathology , T-Lymphocytes/drug effects , Taurine/pharmacology , gamma-Aminobutyric Acid/pharmacology
12.
Front Immunol ; 12: 634749, 2021.
Article in English | MEDLINE | ID: mdl-33679787

ABSTRACT

Childhood vaccines have been the cornerstone tool of public health over the past century. A major barrier to neonatal vaccination is the "immaturity" of the infant immune system and the inefficiency of conventional vaccine approaches at inducing immunity at birth. While much of the literature on fetal and neonatal immunity has focused on the early life propensity toward immune tolerance, recent studies indicate that the fetus is more immunologically capable than previously thought, and can, in some circumstances, mount adaptive B and T cell responses to perinatal pathogens in utero. Although significant hurdles remain before these findings can be translated into vaccines and other protective strategies, they should lend optimism to the prospect that neonatal and even fetal vaccination is achievable. Next steps toward this goal should include efforts to define the conditions for optimal stimulation of infant immune responses, including antigen timing, dose, and route of delivery, as well as antigen presentation pathways and co-stimulatory requirements. A better understanding of these factors will enable optimal deployment of vaccines against malaria and other pathogens to protect infants during their period of greatest vulnerability.


Subject(s)
Fetus/immunology , Immunocompetence , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Adaptive Immunity , Age Factors , Antibodies, Protozoan/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/parasitology , Female , Humans , Immune Tolerance , Immunity, Innate , Immunization Schedule , Infant, Newborn , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/parasitology , Malaria/immunology , Malaria/parasitology , Malaria/transmission , Maternal-Fetal Exchange , Pregnancy , Vaccination
13.
J Biol Chem ; 296: 100523, 2021.
Article in English | MEDLINE | ID: mdl-33711340

ABSTRACT

The Stimulator of Interferon Genes (STING) pathway is implicated in the innate immune response and is important in both oncogenesis and cancer treatment. Specifically, activation of the cytosolic DNA sensor STING in antigen-presenting cells (APCs) induces a type I interferon response and cytokine production that facilitates antitumor immune therapy. However, use of STING agonists (STINGa) as a cancer therapeutic has been limited by unfavorable pharmacological properties and targeting inefficiency due to rapid clearance and limited uptake into the cytosol. Exosomes, a class of extracellular vesicles shed by all cells are under consideration for their use as effective carriers of drugs owing to their innate ability to be taken up by cells and their biocompatibility for optimal drug biodistribution. Therefore, we engineered exosomes to deliver the STING agonist cyclic GMP-AMP (iExoSTINGa), to exploit their favorable pharmacokinetics and pharmacodynamics. Selective targeting of the STING pathway in APCs with iExoSTINGa was associated with superior potency compared with STINGa alone in suppressing B16F10 tumor growth. Moreover, iExoSTINGa showed superior uptake of STINGa into dendritic cells compared with STINGa alone, which led to increased accumulation of activated CD8+ T-cells and an antitumor immune response. Our study highlights the potential of exosomes in general, and iExoSTINGa specifically, in enhancing cancer therapy outcomes.


Subject(s)
Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Exosomes/metabolism , Immunity, Innate/immunology , Melanoma, Experimental/immunology , Membrane Proteins/agonists , Nucleotides, Cyclic/pharmacology , Animals , Antigen-Presenting Cells/drug effects , CD8-Positive T-Lymphocytes/drug effects , Female , HEK293 Cells , Humans , Immunity, Innate/drug effects , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Signal Transduction
14.
Int Immunopharmacol ; 94: 107441, 2021 May.
Article in English | MEDLINE | ID: mdl-33611060

ABSTRACT

Luteolin, a naturally found dietary flavonoid, has a wide range of beneficial biological effects, including effects against tumors and oxidants. Studies proved that luteolin can modulate immune responses. In this study, we investigated the function of luteolin as an antitumor vaccine adjuvant (to treat malignant melanoma) in vitro and in vivo. We found that Luteolin may activated the PI3K-Akt pathways in APCs (Antigen Presenting Cells), induced the activation of APCs, enhanced CTL (Cytotoxic T Lymphocyte) responses, and inhibited tolerogenic T cells. To prove the role of CD8+T cells in immune process, we sorted the CD8+T cells from the immunized mice and transferred them to the B16F10 tumor-bearing mice, the result showed that the survival rate was improved. We also observed that in the mice immunized with Luteolin as an adjuvant, the tumor growth was significantly reduced. Taken together, the result demonstrated that luteolin showed promising properties as a vaccine adjuvant for treating malignant melanoma.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Luteolin/therapeutic use , Melanoma, Experimental/drug therapy , T-Lymphocytes, Cytotoxic/drug effects , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Cancer Vaccines/pharmacology , Cell Line , Disease Models, Animal , Female , Luteolin/pharmacology , Melanoma, Experimental/immunology , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
15.
Sci Rep ; 11(1): 2010, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479373

ABSTRACT

The cell wall of Mycobacterium tuberculosis is composed of diverse glycolipids which potentially interact with the human immune system. To overcome difficulties in obtaining pure compounds from bacterial extracts, we recently synthesized three forms of mycobacterial diacyltrehalose (DAT) that differ in their fatty acid composition, DAT1, DAT2, and DAT3. To study the potential recognition of DATs by human T cells, we treated the lipid-binding antigen presenting molecule CD1b with synthetic DATs and looked for T cells that bound the complex. DAT1- and DAT2-treated CD1b tetramers were recognized by T cells, but DAT3-treated CD1b tetramers were not. A T cell line derived using CD1b-DAT2 tetramers showed that there is no cross-reactivity between DATs in an IFN-γ release assay, suggesting that the chemical structure of the fatty acid at the 3-position determines recognition by T cells. In contrast with the lack of recognition of DAT3 by human T cells, DAT3, but not DAT1 or DAT2, activates Mincle. Thus, we show that the mycobacterial lipid DAT can be both an antigen for T cells and an agonist for the innate Mincle receptor, and that small chemical differences determine recognition by different parts of the immune system.


Subject(s)
Antigens, CD1/genetics , Host-Pathogen Interactions/genetics , Trehalose/genetics , Tuberculosis/enzymology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/pathology , Antigens, CD1/chemistry , Antigens, CD1/immunology , Humans , Interferon-gamma/chemistry , Interferon-gamma/genetics , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lipids/chemistry , Lipids/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Trehalose/chemical synthesis , Trehalose/chemistry , Trehalose/immunology , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/microbiology
16.
Am J Pathol ; 191(4): 720-729, 2021 04.
Article in English | MEDLINE | ID: mdl-33453179

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a widely expressed 50-kDa glycoprotein belonging to the serine protease inhibitor family, with well-established anti-inflammatory functions. Recently, we demonstrated the immunoregulatory role played by PEDF in dry eye disease (DED) by suppressing the maturation of antigen-presenting cells at the ocular surface following exposure to the desiccating stress. In this study, we evaluated the effect of PEDF on the immunosuppressive characteristics of regulatory T cells (Tregs), which are functionally impaired in DED. In the presence of PEDF, the in vitro cultures prevented proinflammatory cytokine (associated with type 17 helper T cells)-induced loss of frequency and suppressive phenotype of Tregs derived from normal mice. Similarly, PEDF maintained the in vitro frequency and enhanced the suppressive phenotype of Tregs derived from DED mice. On systemically treating DED mice with PEDF, moderately higher frequencies and significantly enhanced suppressive function of Tregs were observed in the draining lymphoid tissues, leading to the efficacious amelioration of the disease. Our results demonstrate that PEDF promotes the suppressive capability of Tregs and attenuates their type 17 helper T-cell-mediated dysfunction in DED, thereby playing a role in the suppression of DED.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dry Eye Syndromes/drug therapy , Eye Proteins/pharmacology , Nerve Growth Factors/pharmacology , Serpins/pharmacology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Animals , Antigen-Presenting Cells/drug effects , Cytokines/genetics , Disease Models, Animal , Eye Proteins/metabolism , Female , Mice, Inbred C57BL , Nerve Growth Factors/metabolism , Phenotype , Serpins/metabolism
17.
Nanoscale Horiz ; 6(2): 156-167, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33400743

ABSTRACT

The high mortality associated with glioblastoma multiforme (GBM) is attributed to its invasive nature, hypoxic core, resistant cell subpopulations and a highly immunosuppressive tumor microenvironment (TME). To support adaptive immune function and establish a more robust antitumor immune response, we boosted the local innate immune compartment of GBM using an immunostimulatory mesoporous silica nanoparticle, termed immuno-MSN. The immuno-MSN was specifically designed for systemic and proficient delivery of a potent innate immune agonist to dysfunctional antigen-presenting cells (APCs) in the brain TME. The cargo of the immuno-MSN was cyclic diguanylate monophosphate (cdGMP), a Stimulator of Interferon Gene (STING) agonist. Studies showed the immuno-MSN promoted the uptake of STING agonist by APCs in vitro and the subsequent release of the pro-inflammatory cytokine interferon ß, 6-fold greater than free agonist. In an orthotopic GBM mouse model, systemically administered immuno-MSN particles were taken up by APCs in the near-perivascular regions of the brain tumor with striking efficiency. The immuno-MSNs facilitated the recruitment of dendritic cells and macrophages to the TME while sparing healthy brain tissue and peripheral organs, resulting in elevated circulating CD8+ T cell activity (2.5-fold) and delayed GBM tumor growth. We show that an engineered immunostimulatory nanoparticle can support pro-inflammatory innate immune function in GBM and subsequently augment current immunotherapeutic interventions and improve their therapeutic outcome.


Subject(s)
Brain Neoplasms/therapy , Cyclic GMP/analogs & derivatives , Glioblastoma/therapy , Immunity, Innate/drug effects , Immunologic Factors/therapeutic use , Nanoparticles/therapeutic use , Animals , Antigen-Presenting Cells/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , Cyclic GMP/chemical synthesis , Cyclic GMP/therapeutic use , Dendritic Cells/drug effects , Female , Immunologic Factors/chemical synthesis , Immunotherapy/methods , Interferon Type I/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Porosity , RAW 264.7 Cells , Silicon Dioxide/chemistry , Tumor Microenvironment/drug effects
18.
Cells ; 10(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33430453

ABSTRACT

Gold nanoparticles (AuNPs) have demonstrated outstanding performance in many biomedical applications. Their safety is recognised; however, their effects on the immune system remain ill defined. Antigen-presenting cells (APCs) are immune cells specialised in sensing external stimulus and in capturing exogenous materials then delivering signals for the immune responses. We used primary macrophages (Ms) and dendritic cells (DCs) of mice as an APC model. Whereas AuNPs did not alter significantly Ms and DCs functions, the exposure to AuNPs affected differently Ms and DCs in their responses to subsequent stimulations. The secretion of inflammatory molecules like cytokines (IL-6, TNF-α), chemokine (MCP-1), and reactive oxygen species (ROS) were altered differently in Ms and DCs. Furthermore, the metabolic activity of Ms was affected with the increase of mitochondrial respiration and glycolysis, while only a minor effect was seen on DCs. Antigen presentation to T cells increased when DCs were exposed to AuNPs leading to stronger Th1, Th2, and Th17 responses. In conclusion, our data provide new insights into the complexity of the effects of AuNPs on the immune system. Although AuNPs may be considered as devoid of significant effect, they may induce discrete modifications on some functions that can differ among the immune cells.


Subject(s)
Dendritic Cells/metabolism , Gold/pharmacology , Macrophages/metabolism , Metal Nanoparticles/chemistry , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Biomarkers/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Dendritic Cells/drug effects , Epitopes/drug effects , Glycolysis/drug effects , Gold/toxicity , Macrophages/drug effects , Metal Nanoparticles/toxicity , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Phagocytosis/drug effects , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
19.
Cancer Res ; 81(3): 685-697, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33268528

ABSTRACT

Checkpoint inhibitors (CI) instigate anticancer immunity in many neoplastic diseases, albeit only in a fraction of patients. The clinical success of cyclophosphamide (C)-based haploidentical stem-cell transplants indicates that this drug may re-orchestrate the immune system. Using models of triple-negative breast cancer (TNBC) with different intratumoral immune contexture, we demonstrate that a combinatorial therapy of intermittent C, CI, and vinorelbine activates antigen-presenting cells (APC), and abrogates local and metastatic tumor growth by a T-cell-related effect. Single-cell transcriptome analysis of >50,000 intratumoral immune cells after therapy treatment showed a gene signature suggestive of a change resulting from exposure to a mitogen, ligand, or antigen for which it is specific, as well as APC-to-T-cell adhesion. This transcriptional program also increased intratumoral Tcf1+ stem-like CD8+ T cells and altered the balance between terminally and progenitor-exhausted T cells favoring the latter. Overall, our data support the clinical investigation of this therapy in TNBC. SIGNIFICANCE: A combinatorial therapy in mouse models of breast cancer increases checkpoint inhibition by activating antigen-presenting cells, enhancing intratumoral Tcf1+ stem-like CD8+ T cells, and increasing progenitor exhausted CD8+ T cells.


Subject(s)
Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Cyclophosphamide/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Vinorelbine/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunity, Cellular , Mice , Mice, Inbred BALB C , Transcriptome , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology
20.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188483, 2021 01.
Article in English | MEDLINE | ID: mdl-33232723

ABSTRACT

The upregulation of co-inhibitory checkpoint receptors/ligands that inactivate antitumor T-cells, the enhancement of Tregs-mediated trogocytosis that contribute delayed maturation of antigen presenting cell (APC), and the high Tregs/CD+8 ratio that maintained low threshold of CD+8 cells in the tumor microenvironment (TME); all represent the nuances in the immune evasive strategies of pancreatic ductal adenocarcinoma (PDAC). PDAC is the most aggressive type of pancreatic cancers characterized by poor prognosis and extremely low survivability. Over the years, fraternity of scientists have developed therapeutic agents that can bolster the capacity of the antitumor immunity, usually via the inhibition of immune checkpoints. While this immune checkpoint inhibition therapy represents one major jab from immunity to PDAC, this cancer remains highly resistant due to the acme of desmoplasia in its TME. In this review, we discuss the mechanisms of various checkpoint receptors/ligands axes that are relevant to the fitness of PDAC in its oncogenic ring. These checkpoints include PD-1, CTLA-4, ICOS, TIM-3, TIGIT, BTLA, BTN3A, and VISTA. In addition, we provided evidences that are relevant to the understanding of immune checkpoint inhibition, with extensive outline of immune checkpoint inhibitors that are critical to the treatment of PDAC. Finally, we discuss recently known intricacies of PDAC-mediated immunosuppression, and current advances in treatment options. Having realized that the overall scenario between PDAC and antitumor immunity is like the throwing of jabs in a ring, we therefore discuss future directions and prospect that can knock out PDAC in favor of immunity and humanity.


Subject(s)
B7-H1 Antigen/genetics , CTLA-4 Antigen/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/genetics , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , B7-H1 Antigen/immunology , CTLA-4 Antigen/antagonists & inhibitors , Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Humans , Immunotherapy/methods , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...