Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Mol Immunol ; 20(7): 808-819, 2023 07.
Article in English | MEDLINE | ID: mdl-37225838

ABSTRACT

Innate lymphoid cells (ILCs) are the counterpart of T helper cells in the innate immune system and share multiple phenotypes with T helper cells. Inducible T-cell costimulator (ICOS) is recognized on T cells and participates in T-cell activation and T and B-cell engagement in lymphoid tissues. However, the role of ICOS in ILC3s and ILC3-involved interactions with the immune microenvironment remains unclear. Here, we found that ICOS expression on human ILC3s was correlated with the activated state of ILC3s. ICOS costimulation enhanced the survival, proliferation, and capacity of ILC3s to produce cytokines (IL-22, IL-17A, IFN-γ, TNF, and GM-CSF). Via synergistic effects of ICOS and CD40 signaling, B cells promoted ILC3 functions, and ILC3-induced T-cell-independent B-cell IgA and IgM secretion primarily required CD40 signaling. Hence, ICOS is essential for the nonredundant role of ILC3s and their interaction with adjacent B cells.


Subject(s)
Immunity, Innate , Lymphocytes , Humans , Cytokines , Lymphoid Tissue , Inducible T-Cell Co-Stimulator Protein , B-Lymphocytes
2.
ACS Appl Mater Interfaces ; 15(6): 7878-7886, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36738473

ABSTRACT

Cancer cell membranes (CCMs) are widely used as sources of tumor-associated antigens (TAAs) for the development of cancer vaccines. To improve the CCM-associated cancer vaccine efficiency, personalized cancer vaccines and effective delivery systems are required. In this study, we employed surgically harvested cancer tissues to prepare personalized CCMs for use as TAAs. Thioglycolic-acid-grafted poly(2-methyl-2-oxazoline)-block-poly(2-butyl-2-oxazoline-co-2-butenyl-2-oxazoline) (PMBEOx-COOH) was synthesized to load imiquimod (R837) efficiently. The personalized CCMs were then coated onto R837-loaded PMBEOx-COOH nanoparticles (POxTA NPs/R837) to obtain surgically derived CCM-coated POxTA NPs (SCNPs/R837). SCNPs/R837 efficiently travelled to the draining lymph nodes and were taken up and presented by plasmacytoid dendritic cells to elicit enhanced antitumor immune responses. When combined with programmed cell death-1 antibodies, SCNPs/R837 exhibited high efficiency corresponding to antitumor progression. Therefore, SCNP/R837 might represent a promising personalized cancer vaccine with significant potential for cancer immunotherapy.


Subject(s)
Cancer Vaccines , Nanoparticles , Prostatic Neoplasms , Humans , Male , Imiquimod , Immunotherapy , Antigens, Neoplasm , Dendritic Cells , Prostatic Neoplasms/therapy , Cell Membrane , Cell Line, Tumor
4.
Stem Cell Rev Rep ; 18(7): 2444-2457, 2022 10.
Article in English | MEDLINE | ID: mdl-35274217

ABSTRACT

Mesenchymal stem cells (MSCs) have been used to achieve exciting therapeutic outcomes in many animal studies and clinical trials for various autoimmune diseases, including inflammatory bowel disease (IBD). Type 1 regulatory T (Tr1) cells are the main source of interleukin (IL) 10 in the intestine. Whether Tr1 cells are involved during MSC-mediated IBD treatment is unclear. We treated a murine model of 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis with human umbilical cord-derived MSCs (hUCMSCs) and found that the disease severity was alleviated significantly in a dose-dependent manner. hUCMSCs increased the proportion of Tr1 cells and decreased that of T helper (Th)-1 and Th17 cells in the spleen and mesenteric lymph nodes in different stages of colitis. We found that the upregulation of Tr1 cells by hUCMSCs was abrogated after blocking indoleamine-2,3-dioxygenase (IDO), and IDO knockdown in hUCMSCs reversed the increase in Tr1 cell proportions caused by hUCMSCs in colitis. Moreover, hUCMSCs inhibited apoptosis and promoted the proliferation of Tr1 cells. Our results suggest that Tr1 cells play an important role in the amelioration of IBD by MSCs, and they are the target population for the alleviation of IBD by MSCs, providing meaningful references for the study of therapeutic mechanisms of MSCs in other inflammatory diseases.


Subject(s)
Colitis , Dioxygenases , Inflammatory Bowel Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Colitis/chemically induced , Colitis/therapy , Humans , Inflammatory Bowel Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Mice , T-Lymphocytes, Regulatory/pathology , Trinitrobenzenesulfonic Acid/adverse effects
6.
Front Immunol ; 12: 601611, 2021.
Article in English | MEDLINE | ID: mdl-33708200

ABSTRACT

Background: Innate lymphoid cells (ILCs), so far studied mostly in mouse models, are important tissue-resident innate immune cells that play important roles in the colorectal cancer microenvironment and maintain mucosal tissue homeostasis. Plasmacytoid dendritic cells (pDCs) present complexity in various tumor types and are correlated with poor prognosis. pDCs can promote HIV-1-induced group 3 ILC (ILC3) depletion through the CD95 pathway. However, the role of ILC3s in human colon cancer and their correlation with other immune cells, especially pDCs, remain unclear. Methods: We characterized ILCs and pDCs in the tumor microenvironment of 58 colon cancer patients by flow cytometry and selected three patients for RNA sequencing. Results: ILC3s were negatively correlated, and pDCs were positively correlated, with cancer pathological stage. There was a negative correlation between the numbers of ILC3s and pDCs in tumor tissues. RNA sequencing confirmed the correlations between ILC3s and pDCs and highlighted the potential function of many ILC- and pDC-associated differentially expressed genes in the regulation of tumor immunity. pDCs can induce apoptosis of ILC3s through the CD95 pathway in the tumor-like microenvironment. Conclusions: One of the interactions between ILC3s and pDCs is via the CD95 pathway, which may help explain the role of ILC3s in colon cancer.


Subject(s)
Colonic Neoplasms/immunology , Dendritic Cells/immunology , Lymphocytes/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology , Aged , Colonic Neoplasms/pathology , Dendritic Cells/pathology , Female , Flow Cytometry , Humans , Lymphocytes/pathology , Male , Middle Aged , fas Receptor/immunology
7.
Cell Death Discov ; 7(1): 3, 2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33431816

ABSTRACT

Growth differentiation factor 15 (GDF15), a member of the transforming growth factor ß family, is associated with tumor progression, metastasis, and cell apoptosis. However, controversy persists regarding the role of GDF15 in different tumor types, and its function in glioma stem cells (GSCs) remains unknown. Here, we report that GDF15 promotes the GSC-like phenotype in GSC-like cells (GSCLCs) through the activation of leukemia inhibitor factor (LIF)-STAT3 signaling. Mechanistically, GDF15 was found to upregulate expression of the transcription factor c-Fos, which binds to the LIF promoter, leading to enhanced transcription of LIF in GSCLCs. Furthermore, GDF15 may activate the ERK1/2 signaling pathway in GSCLCs, and the upregulation of LIF expression and the GSC-like phenotype was dependent on ERK1/2 signaling. In addition, the small immunomodulator imiquimod induced GDF15 expression, which in turn activated the LIF-STAT3 pathway and subsequently promoted the GSC-like phenotype in GSCLCs. Thus, our results demonstrate that GDF15 can act as a proliferative and pro-stemness factor for GSCs, and therefore, it may represent a potential therapeutic target in glioma treatment.

8.
Pharmacol Res ; 161: 105293, 2020 11.
Article in English | MEDLINE | ID: mdl-33176206

ABSTRACT

Unmethylated CpG oligodeoxynucleotides (ODNs) activate plasmacytoid dendritic cells (pDCs) and B cells to induce humoral and cellular immunity, and are under development for the treatment of multiple cancers. However, the specific differences in antitumor effects among the three CpG ODN classes when administered as a monotherapy or in co-therapy with the anti-PD-1 antibody are unclear. We compared the immunostimulatory effects in vitro and antitumor effects in a CT26 subcutaneous mouse tumor model among the three CpG ODN classes. We found that CpG-A slightly suppressed tumor growth but possessed no synergistic antitumor effects with the anti-PD-1 antibody. CpG-B at low doses significantly inhibited tumor growth and possessed synergistic antitumor effects with the anti-PD-1 antibody. A high dose of CpG-C was required to achieve antitumor effects comparable to those of CpG-B, which was consistent with the immunostimulatory effects in B-cell proliferation and TLR9-NF-κB activation. Importantly, CpG-C in combination with anti-PD-1 antibody inhibited tumor growth more quickly and effectively than CpG-B because CpG-B significantly upregulated PD-L1 expression on multiple host immune cells to promote tumor immune escape. Moreover, co-therapy increased the infiltration of effector memory T cells. In summary, CpG-B and CpG-C with different optimal concentrations possessed strong antitumor effects, while CpG-C was more rapid and effective for co-therapy with the anti-PD-1 antibody.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/drug therapy , CpG Islands , Immune Checkpoint Inhibitors/pharmacology , Oligodeoxyribonucleotides/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Toll-Like Receptor 9/antagonists & inhibitors , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Synergism , Female , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , Tumor Burden/drug effects , Tumor Escape/drug effects , Tumor Microenvironment
9.
Front Cell Dev Biol ; 8: 262, 2020.
Article in English | MEDLINE | ID: mdl-32391356

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is associated with aggressive clinical cases and poor prognosis despite recent advances in disease treatment. In activated B-cell-like (ABC)-DLBCL, the most severe damaged signaling pathways converge to aberrantly activate the Toll-like receptor (TLR) 7/9/MyD88 pathways, leading to the avoidance of cell death and resistance to chemotherapy. A gain of function mutation in MyD88 (MyD88 L265P) enhanced the NF-κB and JAK-STAT signaling pathways and was associated with dysregulation of TLR signaling in the pathogenesis of ABC-DLBCL. Therefore, inhibition of the TLR signaling network may improve clinical outcomes. In this study, we designed a de novo synthesized oligodeoxynucleotide-based antagonist of TLR7 and TLR9, referred to as HJ901, which competitively binds to TLR7/9. We profiled HJ901 inhibition in various DLBCL cell lines and verified tumor suppression in a xenograft mouse model. We found that HJ901 treatment significantly reduced TLR7- and TLR9-mediated cell proliferation and cytokine production in a time- and dose-dependent manner in various DLBCL cell lines expressing the MyD88 L265P mutation. Moreover, HJ901 prevented tumor growth and downregulated the NF-κB and JAK2-STAT3 signaling pathways in a DLBCL xenograft mouse model with the MyD88 L265P mutation. These results reveal that the anti-tumor effects of the synthesized oligodeoxynucleotide-based antagonist, HJ901, which competitively binds to TLR7/9, may be associated with the downregulation of the NF-κB and JAK2-STAT3 signaling pathways and provide rationale for treating ABC-DLBCL patients with the MyD88 L265P mutation.

10.
Front Pharmacol ; 11: 8, 2020.
Article in English | MEDLINE | ID: mdl-32116691

ABSTRACT

BACKGROUND: C type CpG oligodeoxynucleotides (CpG-C ODNs), possessing the features of both A type and B type CpG ODNs, exert a variety of immunostimulatory activities and have been demonstrated as an effective antitumor immunotherapy. Based on the structural characteristics, we designed 20 potential ODNs with the aim of synthesizing an optimal, novel CpG-C ODN specific to human and murine Toll-like receptor 9 (TLR9). We also sought to investigate the in vitro immunostimulatory and in vivo antitumor effects of the novel CpG-C ODN. METHODS: Twenty potential CpG-C ODNs were screened for their ability to secrete interferon (IFN)-α, and interleukin (IL)-6 and tumor necrosis factor (TNF)-α production for the three most promising sequences were assayed in human peripheral blood mononuclear cells (PBMCs) by enzyme-linked immunosorbent assay (ELISA) or cytometric bead array assay. The functions of human and mouse B cells, and cytokine production in mice induced by the most promising sequence, HP06T07, were determined by flow cytometry and ELISA. Growth and morphology of tumor tissues in in vivo murine models inoculated with CT26 cells were analyzed by a growth inhibition assay and immunohistochemistry, respectively. RESULTS: Among the 20 designed ODNs, HP06T07 significantly induced IFN-α, IL-6, and TNF-α secretion, and promoted B-cell activation and proliferation in a dose-dependent manner in human PBMCs and mouse splenocytes in vitro. Intratumoral injection of HP06T07 notably suppressed tumor growth and prolonged survival in the CT26 subcutaneous mouse model in a dose-dependent manner. HP06T07 administered nine times at 2-day intervals (I2) eradicated tumor growth at both primary and distant sites of CT26 tumors. HP06T07 restrained tumor growth by increasing the infiltration of T cells, NK cells, and plasmacytoid dendritic cells (pDCs). CONCLUSIONS: HP06T07, a novel CpG-C ODN, shows potent immunostimulatory activity in vitro and suppresses tumor growth in the CT26 subcutaneous mouse model.

11.
Materials (Basel) ; 13(1)2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31878171

ABSTRACT

In order to meet the requirements of high-frequency vacuum electronic devices with small size, high current density, and low working temperature, a kind of porous tungsten scandate cathode with micro-blade-type arrays was developed. The micro-blade-type arrays were fabricated by laser engraving technology. Subsequently, the cathode was prepared by a vacuum copper removal process and impregnated with active substances at high temperature. Experimental results show that the cathode exhibits excellent low-temperature electron emission performance and that the maximum pulse electron emission current density reaches 81.18 A/cm2 at 800 °C. The cathode also shows apparent combined thermal-field emission characteristics. Further analysis shows that a high electric field strength plays an important role in the electron emission of the scandate cathode. By virtue of the electric field enhancement effect formed by the fabricated micro-blade-type arrays on the cathode surface, the prepared cathode achieves high electron emission capacity.

12.
J Immunol Res ; 2019: 1325181, 2019.
Article in English | MEDLINE | ID: mdl-31781671

ABSTRACT

Innate lymphoid cells (ILCs) comprise a recently identified subset of innate immune cells that are mainly localized to mucosa-associated tissues. Although they have not yet been fully characterized, they can generally be divided into ILC1s, ILC2s, and ILC3s. ILCs and their corresponding cytokines act as important mediators of the early stages of the immune response during inflammation, tissue repair, and the maintenance of epithelial integrity. Consequently, the dysregulation of ILC subsets might promote inflammation and cancer. Numerous studies have demonstrated that these cells play an important role in maintaining the microecological balance of the small intestine; however, their specific roles in mediating inflammation in this tissue and tumorigenesis remain unclear and controversial. In this review, we focus on recent progress that has helped to gain a better understanding of the role of ILCs in intestinal homeostasis, chronic inflammation, and cancer. Further focused research on the regulation and role of ILCs in intestinal homeostasis and pathology will help to reveal valuable diagnostic and therapeutic targets for the treatment of intestinal diseases.


Subject(s)
Enterocolitis/etiology , Enterocolitis/metabolism , Immunity, Innate , Intestinal Neoplasms/etiology , Intestinal Neoplasms/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Animals , Chronic Disease , Disease Susceptibility , Enterocolitis/pathology , Gastrointestinal Microbiome/immunology , Humans , Immunity, Mucosal , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology
13.
J Immunol Res ; 2019: 1749803, 2019.
Article in English | MEDLINE | ID: mdl-31093508

ABSTRACT

Plasmacytoid dendritic cells (pDCs) express high levels of the toll-like receptors (TLRs) TLR7 and TLR9. In response to TLR7 and TLR9 ligands, pDCs are primary producers of type I interferons. Our previous study demonstrated that pDCs activated by the TLR7 ligand imiquimod (IMQ) and the TLR9 ligand CpG A can kill breast cancer cells in vitro and inhibit tumor growth in vivo. Moreover, we observed a distinctive morphological, phenotypic change in pDCs after activation by IMQ and CpG A. However, the effect of other TLR7 and TLR9 ligands on pDCs remains less understood. In this study, we treat pDCs with the TLR7 ligand IMQ, TLR7/8 ligands (CL097 and CL075), and three TLR9 ligands (different types of CpGs). The size of pDCs increased significantly after activation by TLR7, or TLR7/8 ligands. TLR7, TLR7/8, and TLR9 ligands similarly modulated cytokine release, as well as protein expression of pDC markers, costimulatory molecules, and cytotoxic molecules. Interestingly, TLR7/8 ligands, especially CL097, induced stronger responses. These results are relevant to the further study of the role and mechanism of pDC-induced antitumor effects and may aid in the development of a new strategy for future tumor immunotherapy.


Subject(s)
Cell Differentiation/drug effects , Cytokines/immunology , Dendritic Cells/drug effects , Imiquimod/pharmacology , Membrane Glycoproteins/immunology , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , Animals , Female , Imidazoles/pharmacology , Interferon Inducers/pharmacology , Interferon Type I/immunology , Ligands , Mice , Mice, Inbred BALB C , Quinolines/pharmacology , Thiazoles/pharmacology
14.
Int J Cancer ; 144(11): 2867-2879, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30565657

ABSTRACT

Owing to the limited therapeutic efficacy of glioma vaccines, new strategies are required to improve cancer vaccines. Our study aimed to assess the therapeutic efficacy of a glioma vaccine called STDENVANT. This vaccine, comprising glioma stem-like cell (GSC) lysate, dendritic cells (DCs), and Toll-like receptor (TLR) 9 agonist CpG motif-containing oligodeoxynucleotides (CpG ODNs), was assessed using a GL261-C57BL/6 orthotopic mouse model of glioma. STDENVANT markedly improved survival and tumor regression by enhancing anti-tumor immune function. Moreover, STDENVANT upregulated programmed death 1 (PD-1) and its ligand PD-L1 on effector T cells, DCs, and glioma tissues, resulting in the accumulation of regulatory T (Treg) cells in the brain and lymph nodes. Combinatorial administration of anti-PD-L1 antibody and STDENVANT conferred a greater survival advantage and decreased the Treg cell population in the brain. The present results indicate that PD-L1 blockade can promote tumor regression via STDENVANT in a mouse model of glioma, and combinatorial administration of anti-PD-L1 antibody and STDENVANT increases the therapeutic anti-tumor efficacy of treatment.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Glioma/therapy , Adjuvants, Immunologic/administration & dosage , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Brain/cytology , Brain/immunology , Brain/pathology , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Glioma/immunology , Glioma/mortality , Glioma/pathology , Humans , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/immunology
15.
Front Immunol ; 9: 2491, 2018.
Article in English | MEDLINE | ID: mdl-30443251

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that typically inhibit the translation and stability of messenger RNAs (mRNAs). They are ~22 nucleotides long and control both physiological and pathological processes. Altered expression of miRNAs is often associated with human diseases. Thus, miRNAs have become important therapeutic targets, and some clinical trials investigating the effect of miRNA-based therapeutics in different types of diseases have already been conducted. The tumor microenvironment (TME) comprises cells such as infiltrated immune cells, cancer-associated endothelial cells (CAEs) and cancer-associated fibroblasts (CAFs), and all the components participate in the complicated crosstalk with tumor cells to affect tumor progression. Altered miRNAs expression in both these stromal and tumor cells could drive tumorigenesis. Thus, in this review, we discuss how aberrantly expressed miRNAs influence tumor progression; summarize the crosstalk between infiltrated immune cells, CAEs, CAFs, and tumor cells through miRNAs, and clarify the important roles of miRNAs in the tumor microenvironment, which may facilitate the clinical application of miRNA-based therapies.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Endothelial Cells/immunology , Lymphocytes, Tumor-Infiltrating/immunology , MicroRNAs/genetics , Neoplasms/genetics , Animals , Carcinogenesis , Gene Expression Regulation, Neoplastic , Humans , Immune Tolerance , Immunity, Cellular , Neoplasms/therapy , Tumor Microenvironment/genetics
16.
Anal Bioanal Chem ; 410(8): 2183-2192, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29368150

ABSTRACT

Based on the foaming property of the honey, a rapid, simple, and effective method solvent floatation (SF) was developed and firstly applied to the extraction and separation of triazine herbicides in honey. The analytes were determined by high-performance liquid chromatography. Some parameters affecting the extraction efficiencies, such as the type and volume of extraction solvent, type of salt, amount of (NH4)2SO4, pH value of sample solution, gas flow rate, and floatation time, were investigated and optimized. The limits of detection for analytes are in the range of 0.16-0.56 µg kg-1. The recoveries and relative standard deviations for determining triazines in five real honey samples are in the range of 78.2-112.9 and 0.2-9.2%, respectively.


Subject(s)
Chemical Fractionation/methods , Chromatography, High Pressure Liquid/methods , Herbicides/analysis , Honey/analysis , Triazines/analysis , Chemical Fractionation/instrumentation , Chromatography, High Pressure Liquid/instrumentation , Equipment Design , Food Analysis/instrumentation , Food Analysis/methods , Herbicides/isolation & purification , Ionic Liquids/chemistry , Limit of Detection , Osmolar Concentration , Solvents/chemistry , Triazines/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...