Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 578
Filter
Add more filters

Publication year range
1.
ACS Biomater Sci Eng ; 10(5): 3387-3400, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38656158

ABSTRACT

Given the worldwide problem posed by enteric pathogens, the discovery of safe and efficient intestinal adjuvants combined with novel antigen delivery techniques is essential to the design of mucosal vaccines. In this work, we designed poly (lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs) to codeliver all-trans retinoic acid (atRA), novel antigens, and CpG. To address the insolubility of the intestinal adjuvant atRA, we utilized PLGA to encapsulate atRA and form a "nanocapsid" with polydopamine. By leveraging polydopamine, we adsorbed the water-soluble antigens and the TLR9 agonist CpG onto the NPs' surface, resulting in the pathogen-mimicking PLPCa NPs. In this study, the novel fusion protein (HBf), consisting of the Mycobacterium avium subspecies paratuberculosis antigens HBHA, Ag85B, and Bfra, was coloaded onto the NPs. In vitro, PLPCa NPs were shown to promote the activation and maturation of bone marrow-derived dendritic cells. Additionally, we found that PLPCa NPs created an immune-rich microenvironment at the injection site following intramuscular administration. From the results, the PLPCa NPs induced strong IgA levels in the gut in addition to enhancing powerful systemic immune responses. Consequently, significant declines in the bacterial burden and inflammatory score were noted in PLPCa NPs-treated mice. In summary, PLPCa can serve as a novel and safe vaccine delivery platform against gut pathogens, such as paratuberculosis, capable of activating both systemic and intestinal immunity.


Subject(s)
Nanoparticles , Paratuberculosis , Animals , Nanoparticles/chemistry , Paratuberculosis/immunology , Paratuberculosis/prevention & control , Mice , Tretinoin/chemistry , Tretinoin/pharmacology , Mycobacterium avium subsp. paratuberculosis/immunology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Antigens, Bacterial/immunology , Antigens, Bacterial/chemistry , Dendritic Cells/immunology , Dendritic Cells/drug effects , Intestines/immunology , Intestines/microbiology , Mice, Inbred C57BL , Female , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/administration & dosage , Bacterial Vaccines/immunology , Mice, Inbred BALB C
2.
Biomater Sci ; 12(10): 2672-2688, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38596867

ABSTRACT

Breast cancer, a pervasive malignancy affecting women, demands a diverse treatment approach including chemotherapy, radiotherapy, and surgical interventions. However, the effectiveness of doxorubicin (DOX), a cornerstone in breast cancer therapy, is limited when used as a monotherapy, and concerns about cardiotoxicity persist. Ginsenoside Rg3, a classic compound of traditional Chinese medicine found in Panax ginseng C. A. Mey., possesses diverse pharmacological properties, including cardiovascular protection, immune modulation, and anticancer effects. Ginsenoside Rg3 is considered a promising candidate for enhancing cancer treatment when combined with chemotherapy agents. Nevertheless, the intrinsic challenges of Rg3, such as its poor water solubility and low oral bioavailability, necessitate innovative solutions. Herein, we developed Rg3-PLGA@TMVs by encapsulating Rg3 within PLGA nanoparticles (Rg3-PLGA) and coating them with membranes derived from tumor cell-derived microvesicles (TMVs). Rg3-PLGA@TMVs displayed an array of favorable advantages, including controlled release, prolonged storage stability, high drug loading efficiency and a remarkable ability to activate dendritic cells in vitro. This activation is evident through the augmentation of CD86+CD80+ dendritic cells, along with a reduction in phagocytic activity and acid phosphatase levels. When combined with DOX, the synergistic effect of Rg3-PLGA@TMVs significantly inhibits 4T1 tumor growth and fosters the development of antitumor immunity in tumor-bearing mice. Most notably, this delivery system effectively mitigates the toxic side effects of DOX, particularly those affecting the heart. Overall, Rg3-PLGA@TMVs provide a novel strategy to enhance the efficacy of DOX while simultaneously mitigating its associated toxicities and demonstrate promising potential for the combined chemo-immunotherapy of breast cancer.


Subject(s)
Doxorubicin , Ginsenosides , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Ginsenosides/chemistry , Ginsenosides/pharmacology , Ginsenosides/administration & dosage , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Female , Nanoparticles/chemistry , Mice , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/drug effects , Mice, Inbred BALB C , Cell Line, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Liberation , Drug Carriers/chemistry , Dendritic Cells/drug effects
3.
Int J Pharm ; 656: 124045, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38561134

ABSTRACT

The field of cancer therapy is witnessing the emergence of immunotherapy, an innovative approach that activates the body own immune system to combat cancer. Immunogenic cell death (ICD) has emerged as a prominent research focus in the field of cancer immunotherapy, attracting significant attention in recent years. The activation of ICD can induce the release of damage-associated molecular patterns (DAMPs), such as calreticulin (CRT), adenosine triphosphate (ATP), high mobility group box protein 1 (HMGB1), and heat shock proteins (HSP). Subsequently, this process promotes the maturation of innate immune cells, including dendritic cells (DCs), thereby triggering a T cell-mediated anti-tumor immune response. The activation of the ICD ultimately leads to the development of long-lasting immune responses against tumors. Studies have demonstrated that partial therapeutic approaches, such as chemotherapy with doxorubicin, specific forms of radiotherapy, and phototherapy, can induce the generation of ICD. The main focus of this article is to discuss and review the therapeutic methods triggered by nanoparticles for ICD, while briefly outlining their anti-tumor mechanism. The objective is to provide a comprehensive reference for the widespread application of ICD.


Subject(s)
Immunogenic Cell Death , Immunotherapy , Nanoparticles , Neoplasms , Humans , Immunogenic Cell Death/drug effects , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/drug therapy , Immunotherapy/methods , Animals , Nanoparticles/administration & dosage , Dendritic Cells/immunology , Dendritic Cells/drug effects
4.
Biomater Sci ; 12(9): 2292-2301, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38498328

ABSTRACT

Colorectal cancer (CRC) ranks among the most prevalent cancers globally, demanding innovative therapeutic strategies. Immunotherapy, a promising avenue, employs cancer vaccines to activate the immune system against tumors. However, conventional approaches fall short of eliciting robust responses within the gastrointestinal (GI) tract, where CRC originates. Harnessing the potential of all-trans retinoic acid (ATRA) and cytosine-phosphorothioate-guanine (CpG), we developed layered nanoparticles using a layer-by-layer assembly method to co-deliver these agents. ATRA, crucial for gut immunity, was efficiently encapsulated alongside CpG within these nanoparticles. Administering these ATRA@CpG-NPs, combined with ovalbumin peptide (OVA), effectively inhibited orthotopic CRC growth in mice. Our approach leveraged the inherent benefits of ATRA and CpG, demonstrating superior efficacy in activating dendritic cells, imprinting T cells with gut-homing receptors, and inhibiting tumor growth. This mucosal adjuvant presents a promising strategy for CRC immunotherapy, showcasing the potential for targeting gut-associated immune responses in combating colorectal malignancies.


Subject(s)
Colorectal Neoplasms , Dinucleoside Phosphates , Nanoparticles , Tretinoin , Tretinoin/chemistry , Tretinoin/administration & dosage , Tretinoin/pharmacology , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Mice , Humans , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Mice, Inbred C57BL , Female , Immunotherapy/methods , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovalbumin/chemistry , Cell Line, Tumor , Mice, Inbred BALB C , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Layer-by-Layer Nanoparticles
5.
Molecules ; 27(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35209237

ABSTRACT

Dendritic cells are antigen-presenting cells, which identify and process pathogens to subsequently activate specific T lymphocytes. To regulate the immune responses, DCs have to mature by the recognition of TLR ligands, TNFα or IFNγ. These ligands have been used as adjuvants to activate DCs in situ or in vitro, with toxic effects. It has been shown that some molecules affect the immune system, e.g., Masticadienonic acid (MDA) and 3α-hydroxy masticadienoic acid (3α-OH MDA) triterpenes naturally occurring in several medicinal plants, since they activate the nitric oxide synthase in macrophages and induce T lymphocyte proliferation. The DCs maturation induced by MDA or 3a-OH MDA was determined by incubating these cells with MDA or 3α-OH MDA, and their phenotype was afterwards analyzed. The results showed that only 3α-OH MDA was able to induce DCs maturation. When mice with melanoma were inoculated with DCs/3α-OH MDA, a decreased tumor growth rate was observed along with an extended cell death area within tumors compared to mice treated with DCs incubated with MDA. In conclusion, it is proposed that 3α-OH MDA may be an immunostimulant molecule. Conversely, it is proposed that MDA may be a molecule with anti-inflammatory properties.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Immunomodulation/drug effects , Triterpenes/chemistry , Triterpenes/pharmacology , Animals , Biomarkers , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Immunophenotyping , Mice , Molecular Structure , Xenograft Model Antitumor Assays
6.
J Ethnopharmacol ; 285: 114918, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34919989

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Angelicae pubescentis radix (APR) has a long history in the treatment of rheumatoid arthritis (RA) in China. It has the effects of dispelling wind to eliminate dampness, removing arthralgia and stopping pain in the Chinese Pharmacopeia, but its mechanisms was unclear. Columbianadin (CBN) was one of the main bioactive compounds of APR, and has many pharmacological effects. But the immunosuppressive effect of CBN on DCs and the potential mechanism needed to be explored. AIM OF THE STUDY: The study was aimed to clarify the immunosuppressive effect of CBN on maturation, migration, allogenic T cell stimulation and phagocytosis capacity of TNF-α induced DCs. MATERIALS AND METHODS: Bone marrow-derived DCs were obtained and cultured from C57BL/6 mice in accordance with protocol. The phenotypic study (CD11c, CD40, CD80, CD86 and MHC Ⅱ) were measured by flow cytometry. FITC-dextran were uptaked by DCs and the change of endocytosis activity were mediated by acquired mannose receptor. Transwell chambers were used to detect the migration ability of DCs. Mixed leukocyte reaction (MLR) assay was used to detect the allostimulatory ability of CBN on TNF-α stimulated DCs. The secretion of cytokines and chemokines was measured by ELISA Kit. TLRs gene and MAPKs/NF-κB protein expression were checked by qRT-PCR and Western blot. RESULTS: CBN inhibited the maturation of TNF-α-induced DCs while maintaining phagocytosis capabilities. Additionally, CBN inhibited the migration of TNF-α stimulated DCs, which related to reduce the production of chemokines (MCP-1, MIP-1α). Notably, CBN could suppress the proliferation of CD4+T cells by inhibiting DCs maturation, and decrease the proinflammatory cytokines IL-6 production. Furthermore, CBN inhibited mRNA expression of TLR2, TLR7 and TLR9 in TNF-α-activated DCs. Meanwhile, the phosphorylation of p38, JNK1/2 and NF-κB protein were significantly inhibited in CBN treated DCs. CONCLUSIONS: These findings provided novel insights into the pharmacological activity of CBN. They also indicated that inhibition DCs maturation owning to the immunosuppressive effect of CBN. CBN was expected as a potential immunosuppressant and TLRs/MAPKs/NF-κB pathway may be an important mechanism for CBN's immunosuppressive activity.


Subject(s)
Allogeneic Cells/physiology , Cell Movement/drug effects , Coumarins/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/physiology , T-Lymphocytes/physiology , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phagocytosis , Phytotherapy , Toll-Like Receptors
7.
J Nutr Biochem ; 100: 108880, 2022 02.
Article in English | MEDLINE | ID: mdl-34655755

ABSTRACT

Obesity is associated with the dysregulation of vitamin D metabolism and altered immune responses in bone marrow-derived dendritic cells (BMDCs). Vitamin D can affect the differentiation, maturation, and activation of dendritic cells (DCs) and regulate autophagy via vitamin D receptor signaling. Autophagy was shown to be involved in the functions of DCs. We investigated the effects of dietary vitamin D supplementation and in vitro 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) treatment on autophagy in BMDCs from control diet (CON)-fed lean and high-fat diet (HFD)-induced obese mice. C57BL/6 male mice were fed CON or HFD with 10% or 45% kcal fat, respectively, supplemented with 1,000 or 10,000 IU vitamin D/kg diet (vDC or vDS) for 12 weeks. BMDCs were generated by culturing bone marrow cells from the mice with 20 ng/mL rmGM-CSF and treated with 1 nM 1,25(OH)2D3. Maturation of BMDCs was induced by lipopolysaccharide (50 ng/mL) stimulation. Treatment with 1,25(OH)2D3 inhibited the expression of phenotypes related to DC function (MHC class Ⅱ, CD86, CD80) and production of IL-12p70 by BMDCs from control and obese mice, regardless of dietary vitamin D supplementation. LC3Ⅱ/Ⅰ and VPS34 protein levels increased, and p62 expression decreased, after 1,25(OH)2D3 treatment of the BMDCs in CON-vDC only. Vdr mRNA levels decreased following 1,25(OH)2D3 treatment of BMDCs in the HFD-vDC. In conclusion, autophagy flux was increased by 1,25(OH)2D3 treatment of the BMDCs in CON-vDC but not in the HFD-vDC group. This suggests that the decreased expression of Vdr following 1,25(OH)2D3 treatment might have affected autophagy flux in BMDCs from obese mice.


Subject(s)
Autophagy , Calcitriol/pharmacology , Dendritic Cells/physiology , Diet, High-Fat , Dietary Supplements , Obesity/physiopathology , Vitamin D/administration & dosage , Animals , Bone Marrow Cells/cytology , Dendritic Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Vitamins/administration & dosage
8.
Front Immunol ; 12: 656419, 2021.
Article in English | MEDLINE | ID: mdl-34745081

ABSTRACT

Tuberculosis (TB) is the global health problem with the second highest number of deaths from a communicable disease after COVID-19. Although TB is curable, poor health infrastructure, long and grueling TB treatments have led to the spread of TB pandemic with alarmingly increasing multidrug-resistant (MDR)-TB prevalence. Alternative host modulating therapies can be employed to improve TB drug efficacies or dampen the exaggerated inflammatory responses to improve lung function. Here, we investigated the adjunct therapy of natural immune-modulatory compound berberine in C57BL/6 mouse model of pulmonary TB. Berberine treatment did not affect Mtb growth in axenic cultures; however, it showed increased bacterial killing in primary murine bone marrow-derived macrophages and human monocyte-derived macrophages. Ad libitum berberine administration was beneficial to the host in combination with rifampicin and isoniazid. Berberine adjunctive treatment resulted in decreased lung pathology with no additive or synergistic effects on bacterial burdens in mice. Lung immune cell flow cytometry analysis showed that adjunctive berberine treatment decreased neutrophil, CD11b+ dendritic cell and recruited interstitial macrophage numbers. Late onset of adjunctive berberine treatment resulted in a similar phenotype with consistently reduced numbers of neutrophils both in lungs and the spleen. Together, our results suggest that berberine can be supplemented as an immunomodulatory agent depending on the disease stage and inflammatory status of the host.


Subject(s)
Antitubercular Agents/therapeutic use , Berberine/therapeutic use , Immunologic Factors/therapeutic use , Isoniazid/therapeutic use , Rifampin/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Animals , Antitubercular Agents/pharmacology , Berberine/pharmacology , Cytokines/immunology , Dendritic Cells/drug effects , Drug Therapy, Combination , Female , Humans , Immunologic Factors/pharmacology , Isoniazid/pharmacology , Lung/drug effects , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Neutrophils/drug effects , Neutrophils/immunology , Rifampin/pharmacology , Spleen/drug effects , Spleen/immunology , Spleen/microbiology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
9.
Int Immunopharmacol ; 101(Pt A): 108329, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34749293

ABSTRACT

BACKGROUNDS: Berberine (BBR), a compound long used in traditional Chinese medicine, has been reported to have therapeutic effects in treating ulcerative colitis (UC), attributed to its anti-inflammatory properties and restorative potential of tight junctions (TJs). However, the mechanism by which BBR affects intestinal bacteria and immunity is still unclear. METHODS: This study investigated the effects of BBR on intestinal bacteria and the inflammatory response in dextran sulfate sodium (DSS)-induced colitis mice. Immunohistochemistry (IHC) and electron microscopy were used to detect intestinal TJs. Microflora analysis was used to screen for bacteria regulated by BBR. RESULTS: The results showed that BBR had increased colonic epithelium zonula occludens proteins-1 (ZO-1) and occludin expression and reduced T-helper 17/T regulatory ratio in DSS-induced mice. Mechanically, BBR eliminated DSS-induced intestinal flora disturbances in mice, particularly increased Bacteroides fragilis (B. fragilis) in vivo and in vitro. B. fragilis decreased the interleukin-6 induced by dendritic cells through some heat-resistant component rather than nucleic acids or proteins. CONCLUSIONS: Overall, these data suggest that BBR had a moderating effect on DSS-induced colitis. This compound may regulate intestinal immune cell differentiation by affecting the growth of B. fragilis, providing new insights into the potential application of BBR in UC.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bacteroides fragilis/drug effects , Berberine/pharmacology , Cell Differentiation/drug effects , Colitis/drug therapy , Dendritic Cells/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Bacteroides fragilis/growth & development , Berberine/therapeutic use , Colitis/chemically induced , Colitis, Ulcerative/pathology , Colon/ultrastructure , Cytokines/metabolism , Dextran Sulfate/pharmacology , Flow Cytometry , Gastrointestinal Microbiome/drug effects , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Mice , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction , Tight Junctions/drug effects , Tight Junctions/ultrastructure
10.
Front Immunol ; 12: 732992, 2021.
Article in English | MEDLINE | ID: mdl-34675923

ABSTRACT

Chronic inflammatory disorders (CID), such as autoimmune diseases, are characterized by overactivation of the immune system and loss of immune tolerance. T helper 17 (Th17) cells are strongly associated with the pathogenesis of multiple CID, including psoriasis, rheumatoid arthritis, and inflammatory bowel disease. In line with the increasingly recognized contribution of innate immune cells to the modulation of dendritic cell (DC) function and DC-driven adaptive immune responses, we recently showed that neutrophils are required for DC-driven Th17 cell differentiation from human naive T cells. Consequently, recruitment of neutrophils to inflamed tissues and lymph nodes likely creates a highly inflammatory loop through the induction of Th17 cells that should be intercepted to attenuate disease progression. Tolerogenic therapy via DCs, the central orchestrators of the adaptive immune response, is a promising strategy for the treatment of CID. Tolerogenic DCs could restore immune tolerance by driving the development of regulatory T cells (Tregs) in the periphery. In this review, we discuss the effects of the tolerogenic adjuvants vitamin D3 (VD3), corticosteroids (CS), and retinoic acid (RA) on both DCs and neutrophils and their potential interplay. We briefly summarize how neutrophils shape DC-driven T-cell development in general. We propose that, for optimization of tolerogenic DC therapy for the treatment of CID, both DCs for tolerance induction and the neutrophil inflammatory loop should be targeted while preserving the potential Treg-enhancing effects of neutrophils.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Autoimmune Diseases/drug therapy , Autoimmunity/drug effects , Dendritic Cells/drug effects , Immune Tolerance/drug effects , Inflammation/drug therapy , Neutrophils/drug effects , Th17 Cells/drug effects , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
11.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502035

ABSTRACT

Although fucoidan, a well-studied seaweed-extracted polysaccharide, has shown immune stimulatory effects that elicit anticancer immunity, mucosal adjuvant effects via intranasal administration have not been studied. In this study, the effect of Ecklonia cava-extracted fucoidan (ECF) on the induction of anti-cancer immunity in the lung was examined by intranasal administration. In C57BL/6 and BALB/c mice, intranasal administration of ECF promoted the activation of dendritic cells (DCs), natural killer (NK) cells, and T cells in the mediastinal lymph node (mLN). The ECF-induced NK and T cell activation was mediated by DCs. In addition, intranasal injection with ECF enhanced the anti-PD-L1 antibody-mediated anti-cancer activities against B16 melanoma and CT-26 carcinoma tumor growth in the lungs, which were required cytotoxic T lymphocytes and NK cells. Thus, these data demonstrated that ECF functioned as a mucosal adjuvant that enhanced the immunotherapeutic effect of immune checkpoint inhibitors against metastatic lung cancer.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Laminaria/chemistry , Lung Neoplasms/drug therapy , Polysaccharides/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Combinations , Female , Immune Checkpoint Inhibitors/administration & dosage , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lung Neoplasms/pathology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Plant Extracts , Polysaccharides/administration & dosage , Polysaccharides/pharmacology
12.
J Mater Chem B ; 9(36): 7435-7446, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34551058

ABSTRACT

Cancer vaccines based on DNA encoding oncogenes have shown great potential in preclinical studies. However, the efficacy of DNA vaccines is limited by their weak immunogenicity because of low cellular internalisation and insufficient activation of dendritic cells (DCs). Calcium phosphate (CP) nanoparticles (NPs) are biodegradable vehicles with low toxicity and high loading capacity of DNA but suffer from stability issues. Here we employed adenosine triphosphate (ATP) as a dual functional agent, i.e. stabiliser for CP and immunological adjuvant, and applied the ATP-modified CP (ACP) NPs to the DNA vaccine. ACP NP-enhanced cellular uptake and improved transfection efficiency of DNA vaccine, and further showed the ability to activate DCs that are critical for them to prime T cells in cancer immunotherapy. As a result, a higher level of antigen-specific antibody with stronger tumour growth inhibition was achieved in mice immunised with the ACP-DNA vaccine. Overall, this one-step synthesised ACP NPs are an efficient nano-delivery system and nano-adjuvant for cancer DNA vaccines.


Subject(s)
Adenosine Triphosphate/chemistry , Adjuvants, Immunologic/chemistry , Calcium Phosphates/chemistry , Nanoparticles/chemistry , Vaccines, DNA/chemistry , Animals , Antigen-Antibody Reactions , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Immunotherapy , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Transplantation, Homologous , Vaccination , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
13.
Elife ; 102021 09 02.
Article in English | MEDLINE | ID: mdl-34473049

ABSTRACT

Plasmacytoid dendritic cells (pDCs) constitute a rare type of immune cell with multifaceted functions, but their potential use as a cell-based immunotherapy is challenged by the scarce cell numbers that can be extracted from blood. Here, we systematically investigate culture parameters for generating pDCs from hematopoietic stem and progenitor cells (HSPCs). Using optimized conditions combined with implementation of HSPC pre-expansion, we generate an average of 465 million HSPC-derived pDCs (HSPC-pDCs) starting from 100,000 cord blood-derived HSPCs. Furthermore, we demonstrate that such protocol allows HSPC-pDC generation from whole-blood HSPCs, and these cells display a pDC phenotype and function. Using GMP-compliant medium, we observe a remarkable loss of TLR7/9 responses, which is rescued by ascorbic acid supplementation. Ascorbic acid induces transcriptional signatures associated with pDC-specific innate immune pathways, suggesting an undescribed role of ascorbic acid for pDC functionality. This constitutes the first protocol for generating pDCs from whole blood and lays the foundation for investigating HSPC-pDCs for cell-based immunotherapy.


Subject(s)
Ascorbic Acid/pharmacology , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Dendritic Cells , Hematopoietic Stem Cells , Cells, Cultured , Culture Media/chemistry , Dendritic Cells/cytology , Dendritic Cells/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunotherapy
14.
Nat Commun ; 12(1): 4741, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34362923

ABSTRACT

Biologic therapies have transformed the management of psoriasis, but clinical outcome is variable leaving an unmet clinical need for predictive biomarkers of response. Here we perform in-depth immunomonitoring of blood immune cells of 67 patients with psoriasis, before and during therapy with the anti-TNF drug adalimumab, to identify immune mediators of clinical response and evaluate their predictive value. Enhanced NF-κBp65 phosphorylation, induced by TNF and LPS in type-2 dendritic cells (DC) before therapy, significantly correlates with lack of clinical response after 12 weeks of treatment. The heightened NF-κB activation is linked to increased DC maturation in vitro and frequency of IL-17+ T cells in the blood of non-responders before therapy. Moreover, lesional skin of non-responders contains higher numbers of dermal DC expressing the maturation marker CD83 and producing IL-23, and increased numbers of IL-17+ T cells. Finally, we identify and clinically validate LPS-induced NF-κBp65 phosphorylation before therapy as a predictive biomarker of non-response to adalimumab, with 100% sensitivity and 90.1% specificity in an independent cohort. Our study uncovers important molecular and cellular mediators underpinning adalimumab mechanisms of action in psoriasis and we propose a blood biomarker for predicting clinical outcome.


Subject(s)
Adalimumab/therapeutic use , Dendritic Cells/metabolism , NF-kappa B/metabolism , Psoriasis/immunology , Signal Transduction , B7-H1 Antigen , Biological Therapy , Biomarkers/blood , Dendritic Cells/drug effects , Humans , Interleukin-17 , Lipopolysaccharides/adverse effects , Lymphocytes , Phosphorylation , Sensitivity and Specificity , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factor-alpha
15.
Molecules ; 26(15)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34361596

ABSTRACT

The bitter melon, Momordica charantia L., was once an important food and medicinal herb. Various studies have focused on the potential treatment of stomach disease with M. charantia and on its anti-diabetic properties. However, very little is known about the specific compounds responsible for its anti-inflammatory activities. In addition, the in vitro inhibitory effect of M. charantia on pro-inflammatory cytokine production by lipopolysaccharide (LPS)-stimulated bone marrow-derived dendritic cells (BMDCs) has not been reported. Phytochemical investigation of M. charantia fruit led to the isolation of 15 compounds (1-15). Their chemical structures were elucidated spectroscopically (one- and two-dimensional nuclear magnetic resonance) and with electrospray ionization mass spectrometry. The anti-inflammatory effects of the isolated compounds were evaluated by measuring the production of the pro-inflammatory cytokines interleukin IL-6, IL-12 p40, and tumor necrosis factor α (TNF-α) in LPS-stimulated BMDCs. The cucurbitanes were potent inhibitors of the cytokines TNF-α, IL-6, and IL-12 p40, indicating promising anti-inflammatory effects. Based on these studies and in silico simulations, we determined that the ligand likely docked in the receptors. These results suggest that cucurbitanes from M. charantia are potential candidates for treating inflammatory diseases.


Subject(s)
Bone Marrow Cells/drug effects , Dendritic Cells/drug effects , Fruit/chemistry , Momordica charantia/chemistry , Triterpenes/pharmacology , Animals , Cells, Cultured , Cytokines/metabolism , Mice , Mice, Inbred C57BL
16.
ACS Appl Mater Interfaces ; 13(31): 36824-36838, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34314148

ABSTRACT

Although immuno-oncotherapy in clinic has gained great success, the immunosuppressive tumor microenvironment (TME) existing in the "cold" tumor with insufficient and exhausted lymphocytes may result in a lower-than-expected therapeutic efficiency. Therefore, a properly designed synergistic strategy that can effectively turn the "cold" tumor to "hot" should be considered to improve the therapeutic effects of immuno-oncotherapy. Herein, TME-responsive penetrating nanogels (NGs) were developed, which can improve the delivery and penetration of the co-loaded resiquimod (R848) and green tea catechin (EGCG) in tumors by a nano-sized controlled releasing system of the soluble cyclodextrin-drug inclusion complex. Consequently, the NGs effectively promoted the maturation of dendritic cells, stimulated the cytotoxic T lymphocytes (CTLs), and decreased the PD-L1 expression in tumors. The combination of NGs with the OX40 agonist (αOX40) further synergistically enhanced the activation and infiltration of CTLs into the deep tumor and inhibited the suppression effects from the regulatory T cells (Tregs). As a result, an increased ratio of active CTLs to Tregs in tumors (20.66-fold) was achieved with a 91.56% tumor suppression effect, indicating a successful switch of "cold" tumors to "hot" for an immunologically beneficial TME with significantly improved anti-tumor immune therapeutics. This strategy could be tailored to other immuno-oncotherapeutic approaches to solve the urgent efficiency concerns of the checkpoint-based treatment in clinic.


Subject(s)
Antineoplastic Agents/therapeutic use , Catechin/therapeutic use , Drug Carriers/chemistry , Imidazoles/therapeutic use , Nanogels/chemistry , Neoplasms/drug therapy , 2-Hydroxypropyl-beta-cyclodextrin/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , B7-H1 Antigen/metabolism , Catechin/chemistry , Catechin/pharmacokinetics , Cell Line, Tumor , Dendritic Cells/drug effects , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Hyaluronic Acid/analogs & derivatives , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Immunomodulation , Mice, Inbred C57BL , Neoplasms/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Regulatory/drug effects , Tumor Microenvironment/drug effects
17.
J Ethnopharmacol ; 277: 114256, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34062250

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Herbal polysaccharides have exhibited great immune-enhancing potential. Adjuvants are a key tool for developing efficacious vaccines. In our previous study, a water-soluble polysaccharide extracted from wild Cistanche deserticola Y.C. Ma showed potent immunostimulatory activity. AIM OF STUDY: In this study, the immune profiles and efficacy of aqueous extracts of cultivated Cistanche deserticola Y.C. Ma (AECCD) on ICR mice against ovalbumin (OVA) were investigated. In vitro experiments, the possible DC activation mechanism by AECCD was evaluated. MATERIALS AND METHODS: AECCD were extracted using hot water after which the crude polysaccharides were precipitated by ethanol. Mice were firstly immunized subcutaneously with OVA (10 µg per mouse) alone or OVA (10 µg per mouse) respectively containing different dose of AECCD (200, 400 and 800 µg per mouse) on Days 1 and 14 and the magnitude and kinetics of antibodies and cell-mediated responses were then assessed. RESULTS: AECCD elicited vigorous and long-term IgG responses with mixed Th1/Th2 responses and up-regulated levels of Th-associated cytokines (CD4+IL-4, CD4+IFN-γ and CD8+IFN-γ). Moreover, AECCD induced the strong cellular immune response characterized by increased splenocyte proliferation as well as the activated T cell response. Notably, AECCD significantly enhanced the maturation of dendritic cells (DCs) and inhibited Tregs. In vitro experiments, Preliminary tests indicated that AECCD induced DC activation by promoting phenotypic maturation, cytokine section and allostimulatory activity. Toll-like receptor 4 (TLR4) was an essential receptor for DCs to directly bind AECCD. The inhibitors of NF-κB decreased the expression levels of CD40, CD80, CD86 and MHC-II and the production of IFN-γ, TNF-α and IL-6 through DCs. CONCLUSIONS: Finally, these findings suggested that AECCD could elicit potent and durable antigen specific immune responses through DC activation, which was involved in the regulation of maturation markers and cytokine expression via TLR4-related NF-κB pathway. The study indicates that AECCD is a potential immunomodulator.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cistanche/chemistry , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/isolation & purification , Animals , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Female , Immunologic Factors/administration & dosage , Immunologic Factors/isolation & purification , Immunologic Factors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Ovalbumin , Plant Extracts/administration & dosage , Polysaccharides/administration & dosage , Polysaccharides/isolation & purification
18.
Molecules ; 26(8)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33916909

ABSTRACT

Oxyresveratrol, a stilbene extracted from the plant Artocarpus lakoocha Roxb., has been reported to provide a considerable anti-inflammatory activity. Since the mechanisms of this therapeutic action have been poorly clarified, we investigated whether oxyresveratrol affects the release of the pro-inflammatory cytokines IL-12, IL-6, and TNF-α by human dendritic cells (DCs). We found that oxyresveratrol did not elicit per se the release of these cytokines, but inhibited their secretion induced upon DC stimulation with R848 (Resiquimod), a well-known immune cell activator engaging receptors recognizing RNA viruses. We then investigated whether the inclusion of oxyresveratrol into nanoparticles promoting its ingestion by DCs could favor its effects on cytokine release. For this purpose we synthesized and characterized poly(lactic-co-glycolic acid) (PLGA) nanoparticles, and we assessed their effects on DCs. We found that bare PLGA nanoparticles did not affect cytokine secretion by resting DCs, but increased IL-12, IL-6, and TNF-α secretion by R848-stimulated DCs, an event known as "priming effect". We then loaded PLGA nanoparticles with oxyresveratrol and we observed that oxyresveratrol-bearing particles did not stimulate the cytokine release by resting DCs and inhibited the PLGA-dependent enhancement of IL-12, IL-6, and TNF-α secretion by R848-stimulated DCs. The results herein reported indicate that oxyresveratrol suppresses the cytokine production by activated DCs, thus representing a good anti-inflammatory and immune-suppressive agent. Moreover, its inclusion into PLGA nanoparticles mitigates the pro-inflammatory effects due to cooperation between nanoparticles and R848 in cytokine release. Therefore, oxyresveratrol can be able to contrast the synergistic effects of nanoparticles with microorganisms that could be present in the patient tissues, therefore overcoming a condition unfavorable to the use of some nanoparticles in biological systems.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Imidazoles/adverse effects , Inflammation Mediators/metabolism , Plant Extracts/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer , Stilbenes/administration & dosage , Anti-Inflammatory Agents/chemistry , Cytokines/metabolism , Dendritic Cells/immunology , Drug Carriers/chemistry , Drug Delivery Systems , Drug Synergism , Humans , Nanoparticles/chemistry , Plant Extracts/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Stilbenes/chemistry
19.
Cell Mol Immunol ; 18(5): 1222-1234, 2021 05.
Article in English | MEDLINE | ID: mdl-33767434

ABSTRACT

Aluminum-containing adjuvants have been used for nearly 100 years to enhance immune responses in billions of doses of vaccines. To date, only a few adjuvants have been approved for use in humans, among which aluminum-containing adjuvants are the only ones widely used. However, the medical need for potent and safe adjuvants is currently continuously increasing, especially those triggering cellular immune responses for cytotoxic T lymphocyte activation, which are urgently needed for the development of efficient virus and cancer vaccines. Manganese is an essential micronutrient required for diverse biological activities, but its functions in immunity remain undefined. We previously reported that Mn2+ is important in the host defense against cytosolic dsDNA by facilitating cGAS-STING activation and that Mn2+ alone directly activates cGAS independent of dsDNA, leading to an unconventional catalytic synthesis of 2'3'-cGAMP. Herein, we found that Mn2+ strongly promoted immune responses by facilitating antigen uptake, presentation, and germinal center formation via both cGAS-STING and NLRP3 activation. Accordingly, a colloidal manganese salt (Mn jelly, MnJ) was formulated to act not only as an immune potentiator but also as a delivery system to stimulate humoral and cellular immune responses, inducing antibody production and CD4+/CD8+ T-cell proliferation and activation by either intramuscular or intranasal immunization. When administered intranasally, MnJ also worked as a mucosal adjuvant, inducing high levels of secretory IgA. MnJ showed good adjuvant effects for all tested antigens, including T cell-dependent and T cell-independent antigens, such as bacterial capsular polysaccharides, thus indicating that it is a promising adjuvant candidate.


Subject(s)
Adjuvants, Immunologic/pharmacology , Manganese/pharmacology , Salts/pharmacology , Animals , Antigen Presentation/drug effects , Antiviral Agents/pharmacology , Cancer Vaccines/immunology , Cell Line , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Humans , Interleukin-1/biosynthesis , Interleukin-18/biosynthesis , Macrophages/drug effects , Macrophages/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleotidyltransferases/metabolism , Protein Subunits/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
20.
Med Sci Monit ; 27: e929004, 2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33684094

ABSTRACT

BACKGROUND Selenium and peroxynitrite are known to support the growth and activity of immune cells, including T cells, B cells and macrophages. However, the role of these factors in the immune function of human immature dendritic cells (imDCs) is not clear. MATERIAL AND METHODS Monocytes from a mixture of blood samples were isolated using Ficoll density gradient centrifugation and purified with immunomagnetic beads before being induced into imDCs. Cells then either received no treatment (control group), or treatment with sodium selenite (Na2SeO3, Se), 3-morpholinosydnonimine (SIN1, which decomposes into peroxynitrite), or Se+SIN1. Cell viability, migration, and antiphagocytic abilities, oxidative stress, and protein expression of extracellular signal-regulated kinases (ERK) and MMP2 were assessed using a CCK8 assay, cell counter and flow cytometry, microplate spectrophotometer, and Western blot analysis, respectively. RESULTS Viability of imDCs was unaffected by 0.1 µmol/L of Na2SeO3, although 1 mmol/L of SIN1 decreased it significantly (P<0.05). Chemotactic migration and antiphagocytic abilities were inhibited and enhanced, respectively, by treatment with Na2SeO3 and SIN1 (P<0.05). Activities of superoxide dismutase and glutathione peroxidase were increased by Na2SeO3 and Se+SIN1 (P<0.001). Glutathione content decreased with exposure to Na2SeO3 and SIN1 (P<0.05), but increased after treatment with Se+SIN1 (P<0.05). Levels of reactive oxygen species only increased with SIN1 treatment (P<0.05). Treatment with Na2SeO3, SIN1 and Se+SIN1 increased ERK phosphorylation and decreased MMP2 protein expression (P<0.05). CONCLUSIONS Selenium and peroxynitrite can influence immune function in imDCs by regulating levels of reactive oxygen species or glutathione to activate ERK and promote antigen phagocytosis, as well as by decreasing MMP2 expression to inhibit chemotactic migration.


Subject(s)
Dendritic Cells/drug effects , Peroxynitrous Acid/pharmacology , Selenium/pharmacology , Cell Movement/drug effects , Cell Survival/drug effects , Dendritic Cells/metabolism , Glutathione Peroxidase/metabolism , Humans , Monocytes/drug effects , Monocytes/metabolism , Oxidative Stress/drug effects , Peroxynitrous Acid/immunology , Phagocytosis/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Selenium/immunology , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL