Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.293
Filter
1.
Carbohydr Polym ; 339: 122256, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823922

ABSTRACT

Recently, the intestinal lymphatic transport based on Peyer's patches (PPs) is emerging as a promising absorption pathway for natural polysaccharides. Herein, the aim of this study is to investigate the PP-based oral absorption of a pectic polysaccharide from Smilax china L. (SCLP), as well as its uptake and transport mechanisms in related immune cells. Taking advantages of the traceability of fluorescently labeled SCLP, we confirmed that SCLP could be absorbed into PPs and captured by their mononuclear phagocytes (dendritic cells and macrophages) following oral administration. Subsequently, the systematic in vitro study suggested that the endocytic mechanisms of SCLP by model mononuclear phagocytes (BMDCs and RAW264.7 cells) mainly involved caveolae-mediated endocytosis, macropinocytosis and phagocytosis. More importantly, SCLP directly binds and interacts with toll-like receptor 2 (TLR2) and galectin 3 (Gal-3) receptor, and was taken up by mononuclear phagocytes in receptor-mediated manner. After internalization, SCLP was intracellularly transported primarily through endolysosomal pathway and ultimately localized in lysosomes. In summary, this work reveals novel information and perspectives about the in vivo fate of SCLP, which will contribute to further research and utilization of SCLP and other pectic polysaccharides.


Subject(s)
Peyer's Patches , Smilax , Animals , Mice , RAW 264.7 Cells , Peyer's Patches/metabolism , Smilax/chemistry , Endocytosis , Pectins/chemistry , Pectins/metabolism , Macrophages/metabolism , Macrophages/drug effects , Phagocytosis/drug effects , Phagocytes/metabolism , Phagocytes/drug effects , Toll-Like Receptor 2/metabolism , Mice, Inbred BALB C , Male , Dendritic Cells/metabolism , Dendritic Cells/drug effects , Administration, Oral
2.
Am J Physiol Endocrinol Metab ; 327(1): E55-E68, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38717364

ABSTRACT

Statins are used to treat hypercholesterolemia and function by inhibiting the production of the rate-limiting metabolite mevalonate. As such, statin treatment not only inhibits de novo synthesis of cholesterol but also isoprenoids that are involved in prenylation, the posttranslational lipid modification of proteins. The immunomodulatory effects of statins are broad and often conflicting. Previous work demonstrated that statins increased survival and inhibited myeloid cell trafficking in a murine model of sepsis, but the exact mechanisms underlying this phenomenon were unclear. Herein, we investigated the role of prenylation in chemoattractant responses. We found that simvastatin treatment abolished chemoattractant responses induced by stimulation by C5a and FMLP. The inhibitory effect of simvastatin treatment was unaffected by the addition of either farnesyl pyrophosphate (FPP) or squalene but was reversed by restoring geranylgeranyl pyrophosphate (GGPP). Treatment with prenyltransferase inhibitors showed that the chemoattractant response to both chemoattractants was dependent on geranylgeranylation. Proteomic analysis of C15AlkOPP-prenylated proteins identified several geranylgeranylated proteins involved in chemoattractant responses, including RHOA, RAC1, CDC42, and GNG2. Chemoattractant responses in THP-1 human macrophages were also geranylgeranylation dependent. These studies provide data that help clarify paradoxical findings on the immunomodulatory effects of statins. Furthermore, they establish the role of geranylgeranylation in mediating the morphological response to chemoattractant C5a.NEW & NOTEWORTHY The immunomodulatory effect of prenylation is ill-defined. We investigated the role of prenylation on the chemoattractant response to C5a. Simvastatin treatment inhibits the cytoskeletal remodeling associated with a chemotactic response. We showed that the chemoattractant response to C5a was dependent on geranylgeranylation, and proteomic analysis identified several geranylgeranylated proteins that are involved in C5a receptor signaling and cytoskeletal remodeling. Furthermore, they establish the role of geranylgeranylation in mediating the response to chemoattractant C5a.


Subject(s)
Polyisoprenyl Phosphates , Polyisoprenyl Phosphates/pharmacology , Polyisoprenyl Phosphates/metabolism , Humans , Simvastatin/pharmacology , Chemotactic Factors/pharmacology , Chemotactic Factors/metabolism , Phagocytes/drug effects , Phagocytes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Complement C5a/metabolism , Protein Prenylation/drug effects , Animals , Mice , Sesquiterpenes
3.
Biol Trace Elem Res ; 202(1): 210-220, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37088826

ABSTRACT

In leishmaniasis, the protective immunity is largely mediated by proinflammatory cytokine producing abilities of T cells and an efficient parasite killing by phagocytic cells. Notwithstanding a substantial progress that has been made during last decades, the mechanisms or factors involved in establishing protective immunity against Leishmania are not identified. In ancient Indian literature, metallic "bhasma," particularly that of "swarna" or gold (fine gold particles), is indicated as one of the most prominent metal-based therapeutic medicine, which is known to impart protective and curative properties in various health issues. In this work, we elucidated the potential of swarna bhasma (SB) on the effector properties of phagocytes and antigen-activated CD4+ T cells in augmenting the immunogenicity of L. donovani antigens. The characterization of SB revealing its shape, size, composition, and measurement of cytotoxicity established the physiochemical potential for its utilization as an immunomodulator. The activation of macrophages with SB enhanced their capacity to produce nitric oxide and proinflammatory cytokines, which eventually resulted in reduced uptake of parasites and their proliferation in infected cells. Further, in Leishmania-infected animals, SB administration reduced the generation of IL-10, an anti-inflammatory cytokine, and enhanced pro-inflammatory cytokine generation by antigen activated CD4+ T cells with increased frequency of double (IFNγ+/TNFα+) and triple (IFNγ+TNFα+IL-2+) positive cells and abrogated disease pathogeneses at the early days of infection. Our results also suggested that cow-ghee (A2) emulsified preparation of SB, either alone or with yashtimadhu, a known natural immune modulator which enhances the SB's potential in enhancing the immunogenicity of parasitic antigens. These findings suggested a definite potential of SB in enhancing the effector functions of phagocytes and CD4+ T cells against L. donovani antigens. Therefore, more studies are needed to elucidate the mechanistic details of SB and its potential in enhancing vaccine-induced immunity.


Subject(s)
Antigen Presentation , Antigens, Protozoan , CD4-Positive T-Lymphocytes , Calotropis , Gold , Latex , Leishmania donovani , Macrophages , Medicine, Ayurvedic , Th1 Cells , Arsenic , Drug Combinations , Gold/administration & dosage , Gold/pharmacology , Latex/administration & dosage , Latex/pharmacology , Lead , Macrophages/drug effects , Macrophages/immunology , CD4-Positive T-Lymphocytes/immunology , Phagocytes/drug effects , Phagocytes/immunology , Leishmaniasis/immunology , Leishmaniasis/parasitology , Leishmania donovani/drug effects , Leishmania donovani/growth & development , Leishmania donovani/immunology , Antigens, Protozoan/immunology , Th1 Cells/immunology , Animals , Mice , RAW 264.7 Cells , Female , Mice, Inbred BALB C
4.
Elife ; 112022 02 23.
Article in English | MEDLINE | ID: mdl-35195067

ABSTRACT

The roles of bactericidal cathelicidins against bacterial infection have been extensively studied. However, the antibacterial property and mechanism of action of non-bactericidal cathelicidins are rarely known. Herein, a novel naturally occurring cathelicidin (PopuCATH) from tree frog (Polypedates puerensis) did not't show any direct anti-bacterial activity in vitro. Intriguingly, intraperitoneal injection of PopuCATH before bacterial inoculation significantly reduced the bacterial load in tree frogs and mice, and reduced the inflammatory response induced by bacterial inoculation in mice. PopuCATH pretreatment also increased the survival rates of septic mice induced by a lethal dose of bacterial inoculation or cecal ligation and puncture (CLP). Intraperitoneal injection of PopuCATH significantly drove the leukocyte influx in both frogs and mice. In mice, PopuCATH rapidly drove neutrophil, monocyte/macrophage influx in mouse abdominal cavity and peripheral blood with a negligible impact on T and B lymphocytes, and neutrophils, monocytes/macrophages, but not T and B lymphocytes, were required for the preventive efficacy of PopuCATH. PopuCATH did not directly act as chemoattractant for phagocytes, but PopuCATH obviously drove phagocyte migration when it was cultured with macrophages. PopuCATH significantly elicited chemokine/cytokine production in macrophages through activating p38/ERK mitogen-activated protein kinases (MAPKs) and NF-κB p65. PopuCATH markedly enhanced neutrophil phagocytosis via promoting the release of neutrophil extracellular traps (NETs). Additionally, PopuCATH showed low side effects both in vitro and in vivo. Collectively, PopuCATH acts as a host-based immune defense regulator that provides prophylactic efficacy against bacterial infection without direct antimicrobial effects. Our findings reveal a non-bactericidal cathelicidin which possesses unique anti-bacterial action, and highlight the potential of PopuCATH to prevent bacterial infection.


Subject(s)
Cathelicidins/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages/drug effects , Phagocytes/drug effects , Animals , Anura , Bacteria/drug effects , Bacterial Infections/prevention & control , Bone Marrow Cells , Cathelicidins/chemistry , Cell Line , Chemotaxis , Female , Fungi/drug effects , Humans , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Phagocytes/physiology , Rats
5.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35012988

ABSTRACT

Early diagnosis of oral squamous cell carcinoma (OSCC) remains an unmet clinical need. Therefore, elucidating the initial events of OSCC preceding tumor development could benefit OSCC prognosis. Here, we define the Langerhans cells (LCs) of the tongue and demonstrate that LCs protect the epithelium from carcinogen-induced OSCC by rapidly priming αßT cells capable of eliminating γH2AX+ epithelial cells, whereas γδT and natural killer cells are dispensable. The carcinogen, however, dysregulates the epithelial resident mononuclear phagocytes, reducing LC frequencies, while dendritic cells (DCs), macrophages, and plasmacytoid DCs (pDCs) populate the epithelium. Single-cell RNA-sequencing analysis indicates that these newly differentiated cells display an immunosuppressive phenotype accompanied by an expansion of T regulatory (Treg) cells. Accumulation of the Treg cells was regulated, in part, by pDCs and precedes the formation of visible tumors. This suggests LCs play an early protective role during OSCC, yet the capacity of the carcinogen to dysregulate the differentiation of mononuclear phagocytes facilitates oral carcinogenesis.


Subject(s)
Antineoplastic Agents/metabolism , Carcinogens/toxicity , Langerhans Cells/metabolism , 4-Nitroquinoline-1-oxide/toxicity , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/pathology , Epithelial Cells/metabolism , Epithelium/drug effects , Epithelium/pathology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Histones/metabolism , Humans , Immunity/drug effects , Langerhans Cells/drug effects , Phagocytes/drug effects , Phagocytes/metabolism , Phagocytes/pathology , Quinolones/toxicity , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tongue/pathology , Transcriptome/genetics
6.
Front Immunol ; 12: 770055, 2021.
Article in English | MEDLINE | ID: mdl-34868028

ABSTRACT

Circular RNAs (circRNAs) act as essential regulators in many biological processes, especially in mammalian immune response. Nonetheless, the functions and mechanisms of circRNAs in the invertebrate immune system are largely unclarified. In our previous work, 261 differentially expressed circRNAs potentially related to the development of Apostichopus japonicus skin ulceration syndrome (SUS), which is a major problem restricting the sea cucumber breeding industry, were identified by genome-wide screening. In this study, via miRanda analysis, both circRNA75 and circrRNA72 were shown to share the miR-200 binding site, a key microRNA in the SUS. The two circRNAs were verified to be increased significantly in LPS-exposed primary coelomocytes, similar to the results of circRNA-seq in sea cucumber under Vibrio splendidus-challenged conditions. A dual-luciferase assay indicated that both circRNA75 and circRNA72 could bind miR-200 in vivo, in which circRNA75 had four binding sites of miR-200 and only one for circRNA72. Furthermore, we found that miR-200 could bind the 3'-UTR of Toll interacting protein (Tollip) to negatively mediate the expression of Tollip. Silencing Tollip increased primary coelomocyte apoptosis. Consistently, inference of circRNA75 and circRNA72 could also downregulate Tollip expression, thereby increasing the apoptosis of primary coelomocytes, which could be blocked by miR-200 inhibitor treatment. Moreover, the rate of si-circRNA75-downregulated Tollip expression was higher than that of si-circRNA72 under an equivalent amount. CircRNA75 and circRNA72 suppressed coelomocyte apoptosis by sponging miR-200 to promote Tollip expression. The ability of circRNA to adsorb miRNA might be positively related to the number of binding sites for miRNA.


Subject(s)
Apoptosis/genetics , Digestive System/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , RNA, Circular/genetics , Stichopus/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cells, Cultured , Digestive System/cytology , Digestive System/drug effects , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , Sequence Homology, Nucleic Acid , Stichopus/immunology , Stichopus/virology , Vibrio/immunology , Vibrio/physiology
7.
Cells ; 10(12)2021 11 26.
Article in English | MEDLINE | ID: mdl-34943828

ABSTRACT

Drosophila suzukii is a neobiotic invasive pest that causes extensive damage to fruit crops worldwide. The biological control of this species has been unsuccessful thus far, in part because of its robust cellular innate immune system, including the activity of professional phagocytes known as hemocytes and plasmatocytes. The in vitro cultivation of primary hemocytes isolated from D. suzukii third-instar larvae is a valuable tool for the investigation of hemocyte-derived effector mechanisms against pathogens such as wasp parasitoid larvae, bacteria, fungi and viruses. Here, we describe the morphological characteristics of D. suzukii hemocytes and evaluate early innate immune responses, including extracellular traps released against the entomopathogen Pseudomonas entomophila and lipopolysaccharides. We show for the first time that D. suzukii plasmatocytes cast extracellular traps to combat P. entomophila, along with other cell-mediated reactions, such as phagocytosis and the formation of filopodia.


Subject(s)
Drosophila/immunology , Drosophila/microbiology , Extracellular Traps/metabolism , Immunity, Innate , Introduced Species , Pseudomonas/physiology , Animals , Cell Survival/drug effects , Drosophila/ultrastructure , Extracellular Traps/drug effects , Hemocytes/drug effects , Hemocytes/ultrastructure , Immunity, Innate/drug effects , Larva/cytology , Lipopolysaccharides/pharmacology , Phagocytes/drug effects , Phagocytes/microbiology , Pseudomonas/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism
8.
Front Immunol ; 12: 706727, 2021.
Article in English | MEDLINE | ID: mdl-34777338

ABSTRACT

Tuberculosis (TB) is a significant and continuing problem worldwide, with a death toll of around 1.5 million human lives annually. BCG, the only vaccine against TB, offers a varied degree of protection among human subjects in different regions and races of the world. The majority of the population living near the tropics carries a varying degree of tolerance against BCG due to the widespread prevalence of non-tuberculous mycobacteria (NTM). Interestingly, ≈90% of the Mycobacterium tuberculosis (Mtb) infected population restrain the bacilli on its own, which strengthens the notion of empowering the host immune system to advance the protective efficacy of existing mycobacterial vaccines. In general, Mtb modulates IL-10/STAT3 signaling to skew host mononuclear phagocytes toward an alternatively activated, anti-inflammatory state that helps it thrive against hostile immune advances. We hypothesized that modulating the IL-10/STAT3 driven anti-inflammatory effects in mononuclear cells may improve the prophylactic ability of TB vaccines. This study investigated the immunotherapeutic ability of a porphyrin based small molecule inhibitor of IL-10/STAT3 axis, 5, 15-diphenyl porphyrin (DPP), in improving anti-TB immunity offered by second generation recombinant BCG30 (rBCG30-ARMF-II®) vaccine in mice. The DPP therapy potentiated vaccine induced anti-TB immunity by down-modulating anti-inflammatory responses, while simultaneously up-regulating pro-inflammatory immune effector responses in the immunized host. The employed DPP based immunotherapy led to the predominant activation/proliferation of pro-inflammatory monocytes/macrophages/DCs, the concerted expansion of CD4+/CD8+ effector and central memory T cells, alongside balanced Th17 and Treg cell amplification, and conferred augmented resistance to aerosol Mtb challenge in rBCG30 immunized BALB/c mice.


Subject(s)
BCG Vaccine/immunology , Macrophage Activation/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Porphyrins/pharmacology , Tuberculosis/immunology , Animals , BCG Vaccine/administration & dosage , Cell Plasticity/drug effects , Cytokines/metabolism , Humans , Immunization , Immunomodulation , Immunotherapy , Inflammation Mediators/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Memory T Cells/immunology , Memory T Cells/metabolism , Mice , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tuberculosis/prevention & control
9.
Nat Commun ; 12(1): 6193, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702812

ABSTRACT

Staphylococcus aureus bi-component pore-forming leukocidins are secreted toxins that directly target and lyse immune cells. Intriguingly, one of the leukocidins, Leukocidin AB (LukAB), is found associated with the bacterial cell envelope in addition to secreted into the extracellular milieu. Here, we report that retention of LukAB on the bacterial cells provides S. aureus with a pre-synthesized active toxin that kills immune cells. On the bacteria, LukAB is distributed as discrete foci in two distinct compartments: membrane-proximal and surface-exposed. Through genetic screens, we show that a membrane lipid, lysyl-phosphatidylglycerol (LPG), and lipoteichoic acid (LTA) contribute to LukAB deposition and release. Furthermore, by studying non-covalently surface-bound proteins we discovered that the sorting of additional exoproteins, such as IsaB, Hel, ScaH, and Geh, are also controlled by LPG and LTA. Collectively, our study reveals a multistep secretion system that controls exoprotein storage and protein translocation across the S. aureus cell wall.


Subject(s)
Cell Membrane/metabolism , Cell Wall/metabolism , Staphylococcus aureus/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Cytotoxins/metabolism , Cytotoxins/toxicity , Humans , Leukocidins/metabolism , Leukocidins/toxicity , Lipopolysaccharides/genetics , Lipopolysaccharides/metabolism , Lysine/genetics , Lysine/metabolism , Mice , Phagocytes/drug effects , Phosphatidylglycerols/genetics , Phosphatidylglycerols/metabolism , Protein Transport , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Teichoic Acids/genetics , Teichoic Acids/metabolism , Virulence Factors/toxicity
10.
Cell ; 184(21): 5338-5356.e21, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34624222

ABSTRACT

The tumor microenvironment (TME) influences cancer progression and therapy response. Therefore, understanding what regulates the TME immune compartment is vital. Here we show that microbiota signals program mononuclear phagocytes in the TME toward immunostimulatory monocytes and dendritic cells (DCs). Single-cell RNA sequencing revealed that absence of microbiota skews the TME toward pro-tumorigenic macrophages. Mechanistically, we show that microbiota-derived stimulator of interferon genes (STING) agonists induce type I interferon (IFN-I) production by intratumoral monocytes to regulate macrophage polarization and natural killer (NK) cell-DC crosstalk. Microbiota modulation with a high-fiber diet triggered the intratumoral IFN-I-NK cell-DC axis and improved the efficacy of immune checkpoint blockade (ICB). We validated our findings in individuals with melanoma treated with ICB and showed that the predicted intratumoral IFN-I and immune compositional differences between responder and non-responder individuals can be transferred by fecal microbiota transplantation. Our study uncovers a mechanistic link between the microbiota and the innate TME that can be harnessed to improve cancer therapies.


Subject(s)
Interferon Type I/metabolism , Membrane Proteins/metabolism , Microbiota , Monocytes/metabolism , Tumor Microenvironment , Akkermansia/drug effects , Akkermansia/physiology , Animals , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dietary Fiber/pharmacology , Dinucleoside Phosphates/administration & dosage , Dinucleoside Phosphates/pharmacology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunomodulation/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Macrophages/drug effects , Macrophages/metabolism , Melanoma/immunology , Melanoma/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbiota/drug effects , Monocytes/drug effects , Phagocytes/drug effects , Phagocytes/metabolism , Transcription, Genetic/drug effects , Tumor Microenvironment/drug effects
11.
Sci Rep ; 11(1): 17958, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504130

ABSTRACT

Several plant lectins, or carbohydrate-binding proteins, interact with glycan moieties on the surface of immune cells, thereby influencing the immune response of these cells. Orysata, a mannose-binding lectin from rice, has been reported to exert immunomodulatory activities on insect cells. While the natural lectin is non-glycosylated, recombinant Orysata produced in the yeast Pichia pastoris (YOry) is modified with a hyper-mannosylated N-glycan. Since it is unclear whether this glycosylation can affect the YOry activity, non-glycosylated rOrysata was produced in Escherichia coli (BOry). In a comparative analysis, both recombinant Orysata proteins were tested for their carbohydrate specificity on a glycan array, followed by the investigation of the carbohydrate-dependent agglutination of red blood cells (RBCs) and the carbohydrate-independent immune responses in Drosophila melanogaster S2 cells. Although YOry and BOry showed a similar carbohydrate-binding profiles, lower concentration of BOry were sufficient for the agglutination of RBCs and BOry induced stronger immune responses in S2 cells. The data are discussed in relation to different hypotheses explaining the weaker responses of glycosylated YOry. In conclusion, these observations contribute to the understanding how post-translational modification can affect protein function, and provide guidance in the selection of the proper expression system for the recombinant production of lectins.


Subject(s)
Drosophila melanogaster/cytology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Mannose-Binding Lectins/metabolism , Mannose-Binding Lectins/pharmacology , Oryza/chemistry , Phagocytes/drug effects , Phagocytes/immunology , Plant Lectins/metabolism , Plant Lectins/pharmacology , Polysaccharides/metabolism , Animals , Cell Line , Erythrocytes/drug effects , Erythrocytes/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Glycosylation , Hemagglutination/drug effects , Mannose-Binding Lectins/genetics , Phagocytes/metabolism , Plant Lectins/genetics , Protein Binding , Rabbits , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Saccharomycetales/genetics , Saccharomycetales/metabolism
12.
ACS Appl Mater Interfaces ; 13(34): 40415-40428, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34470103

ABSTRACT

Toxoplasma gondii (T. gondii) infection causes severe zoonotic toxoplasmosis, which threatens the safety of almost one-third of the human population globally. However, there is no effective protective vaccine against human toxoplasmosis. This necessitates anti-T. gondii vaccine development, which is a main priority of public health. In this study, we optimized the adjuvant system 04 (AS04), a vaccine adjuvant constituted by 3-O-desacyl-4'-monophosphoryl lipid A (a TLR4 agonist) and aluminum salts, by packing it within natural extracts of ß-glucan particles (GPs) from Saccharomyces cerevisiae to form a GP-AS04 hybrid adjuvant system. Through a simple mixing procedure, we loaded GP-AS04 particles with the total extract (TE) of T. gondii lysate, forming a novel anti-T. gondii vaccine GP-AS04-TE. Results indicated that the hybrid adjuvant can efficiently and stably load antigens, mediate antigen delivery, facilitate the dendritic uptake of antigens, boost dendritic cell maturation and stimulation, and increase the secretion of pro-inflammatory cytokines. In the mouse inoculation model, GP-AS04-TE significantly stimulated the function of dendritic cells, induced a very strong TE-specific humoral and cellular immune response, and finally showed a strong and effective protection against toxoplasma chronic and acute infections. This work proves the potential of GP-AS04 for exploitation as a vaccine against a range of pathogens.


Subject(s)
Adjuvants, Vaccine/therapeutic use , Aluminum Hydroxide/therapeutic use , Lipid A/analogs & derivatives , Nanocomposites/therapeutic use , Protozoan Vaccines/therapeutic use , Toxoplasma/immunology , Toxoplasmosis/prevention & control , Adjuvants, Vaccine/chemistry , Adjuvants, Vaccine/toxicity , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/immunology , Aluminum Hydroxide/toxicity , Animals , Dendritic Cells/drug effects , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/therapeutic use , Fungal Polysaccharides/toxicity , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Lipid A/chemistry , Lipid A/immunology , Lipid A/therapeutic use , Lipid A/toxicity , Male , Mice, Inbred C57BL , Nanocomposites/chemistry , Nanocomposites/toxicity , Phagocytes/drug effects , Protozoan Vaccines/chemistry , Protozoan Vaccines/immunology , Protozoan Vaccines/toxicity , Saccharomyces cerevisiae/chemistry , Tissue Extracts/chemistry , Tissue Extracts/immunology , Tissue Extracts/therapeutic use , Tissue Extracts/toxicity , Toxoplasma/chemistry , Toxoplasmosis/immunology , beta-Glucans/chemistry , beta-Glucans/therapeutic use , beta-Glucans/toxicity
13.
Int Immunopharmacol ; 99: 108068, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34426114

ABSTRACT

Oligomannose-coated liposomes (OMLs) comprised of dipalmitoylphosphatidylcholine, cholesterol and Man3-DPPE at a molar ratio of 1:1:0.1 and particle diameters of about 1000 nm can induce liposome-encased antigen-specific strong Th1 immunity. In this study, we evaluated the effect of particle sizes of OMLs on induction of Th1 immune responses in mice. Spleen cells obtained from mice immunized with antigen-encapsulating OMLs with 1000- and 800-nm diameters secreted remarkably high levels of IFN-γ upon in vitro stimulation. In addition, sera of mice that received these OMLs had significantly higher titers of antigen-specific IgG2a than those of IgG1, which are commonly associated with Th1 responses. In contrast, treatment with antigen-encapsulating OMLs with 400- and 200-nm diameters failed to induce IFN-γ secretion from spleen cells, although these OMLs did elicit elevation of antigen-specific IgGs. In addition, the titers of serum antigen-specific IgG2a were the same as those of IgG1 in mice that received 400-nm OMLs. Resident peritoneal mononuclear phagocytes (MNPs) treated with OMLs of diameter ≥ 600 nm secreted IL-12, which is essential for induction of Th1 immune responses, while those treated with OMLs of ≤ 400 nm failed to produce this cytokine. However, 400-nm OMLs did induce enhanced expression of MHC class II and costimulatory molecules on MNPs, similarly to OMLs of ≥ 600 nm. Taken together, these results strongly indicate that OMLs of diameter ≥ 600 nm are required to induce Th1 immune responses against OML-encased antigens, although OMLs of diameter ≤ 400 nm can activate MNPs.


Subject(s)
Liposomes/chemistry , Liposomes/immunology , Mannose/chemistry , Mannose/immunology , Th1 Cells/immunology , 1,2-Dipalmitoylphosphatidylcholine/immunology , Animals , Antigens/immunology , B7-2 Antigen/metabolism , Cytochalasin D/pharmacology , Female , Histocompatibility Antigens Class II/drug effects , Histocompatibility Antigens Class II/metabolism , Immune System , Immunoglobulin G/blood , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-12 Subunit p35/metabolism , Mice , Particle Size , Peritoneal Absorption/drug effects , Phagocytes/drug effects , Phagocytes/metabolism , Phagocytosis/drug effects , Spleen/drug effects , Spleen/metabolism
14.
J Immunol ; 207(2): 483-492, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34193599

ABSTRACT

Alcohol use disorders (AUD) increase susceptibility to respiratory infections by 2- to 4-fold in part because of impaired alveolar macrophage (AM) immune function. Alcohol causes AM oxidative stress, diminishing AM phagocytic capacity and clearance of microbes from the alveolar space. Alcohol increases AM NADPH oxidases (Noxes), primary sources of AM oxidative stress, and reduces peroxisome proliferator-activated receptor γ (PPARγ) expression, a critical regulator of AM immune function. To investigate the underlying mechanisms of these alcohol-induced AM derangements, we hypothesized that alcohol stimulates CCAAT/enhancer-binding protein ß (C/EBPß) to suppress Nox-related microRNAs (miRs), thereby enhancing AM Nox expression, oxidative stress, and phagocytic dysfunction. Furthermore, we postulated that pharmacologic PPARγ activation with pioglitazone would inhibit C/EBPß and attenuate alcohol-induced AM dysfunction. AM isolated from human AUD subjects or otherwise healthy control subjects were examined. Compared with control AM, alcohol activated AM C/EBPß, decreased Nox1-related miR-1264 and Nox2-related miR-107, and increased Nox1, Nox2, and Nox4 expression and activity. These alcohol-induced AM derangements were abrogated by inhibition of C/EBPß, overexpression of miR-1264 or miR-107, or pioglitazone treatment. These findings define novel molecular mechanisms of alcohol-induced AM dysfunction mediated by C/EBPß and Nox-related miRs that are amenable to therapeutic targeting with PPARγ ligands. These results demonstrate that PPARγ ligands provide a novel and rapidly translatable strategy to mitigate susceptibility to respiratory infections and related morbidity in individuals with AUD.


Subject(s)
Alcoholism/drug therapy , Alcoholism/metabolism , Ethanol/adverse effects , Macrophages, Alveolar/drug effects , Phagocytes/drug effects , Pioglitazone/pharmacology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Humans , Lung/drug effects , Lung/metabolism , Macrophages, Alveolar/metabolism , Male , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , PPAR gamma/metabolism , Phagocytes/metabolism
15.
Exp Cell Res ; 405(2): 112709, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34174318

ABSTRACT

We earlier identified native human trabecular meshwork stem cells (TMSCs) based on two-parameters- high ABCG2 expression and high nucleus to cytoplasmic ratio. The TMSCs also expressed p75 and AnkyrinG. Based on the high expression of ABCG2 and p75, the TMSCs were identified to be located in the Schwalbe's line region of the TM. In continuation, the current study aimed at elucidating the functional characteristics of human TMSCs. Upon culturing, only a small proportion of TM cells (0.96 ± 0.21% in <30 years) expressing stem cell markers ABCG2 and p75 adhered to the culture dish. This proportion significantly reduced with ageing (0.32 ± 0.23% in 30-60 years and 0.35 ± 0.04% in >60 years). Characterization of the primary TM cultures identified 7.00 ± 1.80% of stem cells with label retaining property. Further, cultured cells had the ability to form TM spheres (0.82 ± 0.23%) which consisted of high ABCG2 and p75 positive cells. Upon dexamethasone induction, 86.00 ± 14.87% and 64.60 ± 7.24% of the cells derived from the TM spheres expressed myocilin and exhibited cross linked actin networks respectively, indicating differentiation of the TMSCs in the sphere to TM cells. In addition, the sphere derived TM cells also possessed phagocytic potential (13.28 ± 3.30%) equivalent to primary TM cells (16.33 ± 4.04%) which was evident upon internalization of zymosan particles. In conclusion, this study has established that a proportion of cultured TM cells had the label retaining property as well as sphere forming ability of adult stem cells. Thus, these results confirm the presence of adult stem cells in the human TM that might be responsible for the maintenance of tissue homeostasis.


Subject(s)
Cell Differentiation/drug effects , Dexamethasone/pharmacology , Stem Cells/drug effects , Trabecular Meshwork/drug effects , Adult , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glycoproteins/metabolism , Homeostasis/physiology , Humans , Phagocytes/cytology , Phagocytes/drug effects , Stem Cells/cytology , Trabecular Meshwork/metabolism
16.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33986194

ABSTRACT

Chagas disease is caused by infection with the protozoan parasite Trypanosoma cruzi (T. cruzi), an intracellular pathogen that causes significant morbidity and death among millions in the Americas from Canada to Argentina. Current therapy involves oral administration of the nitroimidazole benznidazole (BNZ), which has serious side effects that often necessitate cessation of treatment. To both avoid off-target side effects and reduce the necessary dosage of BNZ, we packaged the drug within poly(ethylene glycol)-block-poly(propylene sulfide) polymersomes (BNZ-PSs). We show that these vesicular nanocarriers enhanced intracellular delivery to phagocytic cells and tested this formulation in a mouse model of T. cruzi infection. BNZ-PS is not only nontoxic but also significantly more potent than free BNZ, effectively reducing parasitemia, intracellular infection, and tissue parasitosis at a 466-fold lower dose of BNZ. We conclude that BNZ-PS was superior to BNZ for treatment of T. cruzi infection in mice and that further modifications of this nanocarrier formulation could lead to a wide range of custom controlled delivery applications for improved treatment of Chagas disease in humans.


Subject(s)
Chagas Disease/drug therapy , Nanoparticle Drug Delivery System , Nitroimidazoles/administration & dosage , Phagocytes/parasitology , Trypanocidal Agents/administration & dosage , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Carriers , Mice , Nitroimidazoles/pharmacology , Phagocytes/drug effects , Polyethylene Glycols , Sulfides , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects
17.
Mol Pharm ; 18(6): 2406-2415, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33896187

ABSTRACT

Gangliosides (glycosphingolipids) reduce antibody production by inhibiting B-cell receptor (BCR) signaling. We have shown that a copresentation of gangliosides and polyethylene glycol (PEG) on the same liposomes suppresses anti-PEG IgM production in mice. In addition, we recently observed that pDNA incorporated in PEGylated cationic liposomes (PCLs) induces anti-DNA IgM, which could be a hurdle to the development of efficient gene delivery systems. Therefore, the focus of this study was to determine if the copresentation of gangliosides and DNA on the same PCL would suppress antibody production against DNA. PCLs including DNA induced both anti-PEG IgM production and anti-DNA IgM production. The extent of anti-PEG and anti-DNA IgM production was likely dependent on the immunogenicity of the complexed DNA. Treatment of clodronate-containing liposomes, which causes a depletion of phagocytic cells, suppressed anti-PEG IgM production from PCLs that did not include DNA but failed to suppress anti-PEG IgM production from PCLs that complexed DNA (PCLD). Both anti-PEG IgM and anti-DNA IgM was induced in T-cell-deficient nude mice as well as in normal mice following treatment with PCLs and PCLD, respectively. These results indicate that phagocytic cells contribute to anti-PEG IgM production but not to anti-DNA IgM production, while T-cells do not contribute to any form of antibody production. The copresentation of gangliosides and DNA significantly reduced anti-PEG IgM production but unfortunately did not reduce anti-DNA IgM production. It appears that the immunosuppressive effect of gangliosides, presumably via the CD22 signaling pathway, is limited only to anti-PEG immunity.


Subject(s)
Clodronic Acid/administration & dosage , DNA/immunology , Gangliosides/immunology , Gene Transfer Techniques/adverse effects , Immunoglobulin M/metabolism , Animals , Antibody Formation , Cations , Gangliosides/chemistry , Genetic Therapy/methods , Liposomes , Male , Mice , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , Plasmids/administration & dosage , Plasmids/genetics , Polyethylene Glycols/chemistry
18.
Med Mycol ; 59(8): 749-762, 2021 Jul 14.
Article in English | MEDLINE | ID: mdl-33550415

ABSTRACT

Paracoccidioides brasiliensis is the major etiologic agent of Paracoccidioidomycosis (PCM), the most frequent human deep mycosis in Latin America. It is proposed that masking of ß-glucan in P. brasiliensis cell wall is a critical virulence factor that contributes to the development of a chronic disease characterized by a long period of treatment, which is usually toxic. In this context, the search for immunomodulatory agents for therapeutic purposes is highly desirable. One strategy is to use pattern recognition receptors (PRRs) ligands to stimulate the immune response mediated by phagocytes. Here, we sought to evaluate if Zymosan, a ß-glucan-containing ligand of the PRRs Dectin-1/TLR-2, would enhance phagocyte function and the immune response of mice challenged with P. brasiliensis. Dendritic cells (DCs) infected with P. brasiliensis and treated with Zymosan showed improved secretion of several proinflammatory cytokines and expression of maturation markers. In addition, when cocultured with splenic lymphocytes, these cells induced the production of a potential protective type 1 and 17 cytokine patterns. In macrophages, Zymosan ensued a significant fungicidal activity associated with nitric oxide production and phagolysosome acidification. Importantly, we observed a protective effect of Zymosan-primed DCs delivered intranasally in experimental pulmonary PCM. Overall, our findings support the potential use of ß-glucan-containing compounds such as Zymosan as an alternative or complementary antifungal therapy. LAY SUMMARY: We report for the first time that Paracoccidioides brasiliensis-infected phagocytes treated with Zymosan (cell wall extract from bakers' yeast) show enhanced cytokine production, maturation, and fungal killing. Also, Zymosan-primed phagocytes induce a protective immune response in infected mice.


Subject(s)
Paracoccidioides/immunology , Paracoccidioidomycosis/drug therapy , Phagocytes/drug effects , Zymosan/pharmacology , Animals , Mice , Paracoccidioides/pathogenicity , Paracoccidioidomycosis/immunology , Phagocytes/immunology , Virulence , Zymosan/therapeutic use
19.
RNA Biol ; 18(5): 604-618, 2021 05.
Article in English | MEDLINE | ID: mdl-33622174

ABSTRACT

A persisting obstacle in human immunology is that blood-derived leukocytes are notoriously difficult to manipulate at the RNA level. Therefore, our knowledge about immune-regulatory RNA-networks is largely based on tumour cell-line and rodent knockout models, which do not fully mimic human leukocyte biology. Here, we exploit straightforward cell penetrating peptide (CPP) chemistry to enable efficient loss-of-function phenotyping of regulatory RNAs in primary human blood-derived cells. The classical CPP octaarginine (R8) enabled antisense peptide-nucleic-acid (PNA) oligomer delivery into nearly 100% of human blood-derived macrophages without apparent cytotoxicity even up to micromolar concentrations. In a proof-of-principle experiment, we successfully de-repressed the global microRNA-155 regulome in primary human macrophages using a PNA-R8 oligomer, which phenocopies a CRISPR-Cas9 induced gene knockout. Interestingly, although it is often believed that fairly high concentrations (µM) are needed to achieve antisense activity, our PNA-R8 was effective at 200 nM. RNA-seq characterized microRNA-155 as a broad-acting riboregulator, feedback restraining a late myeloid differentiation-induced pro-inflammatory network, comprising MyD88-signalling and ubiquitin-proteasome components. Our results highlight the important role of the microRNA machinery in fine-control of blood-derived human phagocyte immunity and open the door for further studies on regulatory RNAs in difficult-to-transfect primary human immune cells.


Subject(s)
Inflammation/genetics , MicroRNAs/physiology , Oligonucleotides, Antisense/pharmacology , Phagocytes/drug effects , Cells, Cultured , Gene Knockdown Techniques , Humans , Inflammation/metabolism , MicroRNAs/genetics , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloid Cells/physiology , Phagocytes/immunology , Phagocytes/metabolism , Primary Cell Culture , RNA Interference/drug effects , Transcriptome/drug effects , U937 Cells
20.
Theranostics ; 11(5): 2232-2246, 2021.
Article in English | MEDLINE | ID: mdl-33500722

ABSTRACT

Rationale: Acute lung injury (ALI)-recruited mononuclear phagocytes play a pivotal role in lung injury and repair. This study investigated the types of recruited mononuclear phagocytes and the immunotherapeutic effects of allograft mesenchymal stem cells (MSCs) in a mouse model of lipopolysaccharide (LPS)-induced ALI. Methods: C57BL/6 mice were orotracheally instilled with LPS (20 mg/kg). Compact bone-derived MSCs were administered orotracheally 4 h after LPS inhalation. Mononuclear phagocytes recruited in the lung tissues were characterized at different timepoints by high-dimensional analysis including flow cytometry, mass cytometry, and single-cell RNA sequencing. Results: Eight mononuclear phagocyte subsets recruited to LPS-challenged lungs were precisely identified. On day 3 after LPS administration, both Ly6ChiCD38+ and Ly6ClowCD38+ monocytes were recruited into acutely injured lungs, which was associated with increased secretion of neutrophil chemokines. Ly6ChiCD38+ monocytes differentiated into M1 macrophages on day 3, and subsequently differentiated into CD38+ monocyte-derived dendritic cells (mo-DCs) on day 7, while Ly6ClowCD38+ monocytes differentiated into CD11b+CD38+ DCs on day 7. When ALI mice were treated with MSCs, the mortality significantly reduced. Notably, MSCs reduced the amount of M1 macrophages and reduced the secretion of neutrophil chemokines on day 3. Furthermore, MSCs reduced the number of CD38+ mo-DCs and CD11b+CD38+ DCs on day 7, suppressing the antigen presentation process. Recruited mononuclear phagocyte subsets with a high level of CD38 exhibited an activated phenotype and could secrete higher levels of cytokines and chemokines. Conclusions: This study characterized the dynamic functions and phenotypes of recruited mononuclear phagocytes in ALI mice and MSC-treated ALI mice.


Subject(s)
Acute Lung Injury/therapy , Disease Models, Animal , Immunomodulation , Macrophages/immunology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Cell Differentiation , Cytokines/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/immunology , Phagocytes/drug effects , Phagocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...