Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 502
Filter
1.
Blood ; 143(16): 1599-1615, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38394668

ABSTRACT

ABSTRACT: Treatment resistance of leukemia stem cells (LSCs) and suppression of the autologous immune system represent major challenges to achieve a cure in acute myeloid leukemia (AML). Although AML blasts generally retain high levels of surface CD38 (CD38pos), LSCs are frequently enriched in the CD34posCD38neg blast fraction. Here, we report that interferon gamma (IFN-γ) reduces LSCs clonogenic activity and induces CD38 upregulation in both CD38pos and CD38neg LSC-enriched blasts. IFN-γ-induced CD38 upregulation depends on interferon regulatory factor 1 transcriptional activation of the CD38 promoter. To leverage this observation, we created a novel compact, single-chain CD38-CD3 T-cell engager (BN-CD38) designed to promote an effective immunological synapse between CD38pos AML cells and both CD8pos and CD4pos T cells. We demonstrate that BN-CD38 engages autologous CD4pos and CD8pos T cells and CD38pos AML blasts, leading to T-cell activation and expansion and to the elimination of leukemia cells in an autologous setting. Importantly, BN-CD38 engagement induces the release of high levels of IFN-γ, driving the expression of CD38 on CD34posCD38neg LSC-enriched blasts and their subsequent elimination. Critically, although BN-CD38 showed significant in vivo efficacy across multiple disseminated AML cell lines and patient-derived xenograft models, it did not affect normal hematopoietic stem cell clonogenicity and the development of multilineage human immune cells in CD34pos humanized mice. Taken together, this study provides important insights to target and eliminate AML LSCs.


Subject(s)
Interferon-gamma , Leukemia, Myeloid, Acute , T-Lymphocytes , Animals , Humans , Mice , ADP-ribosyl Cyclase 1/immunology , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Cell Line, Tumor , Hematopoietic Stem Cells/metabolism , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Lymphocyte Activation/drug effects
2.
EBioMedicine ; 87: 104420, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36592514

ABSTRACT

BACKGROUND: Whether immunotherapy combined with different histone deacetylases (HDAC) inhibitors in refractory or relapsed natural killer/T-cell lymphoma (NKTCL) is superior to each agent is still lacking in head-to-head clinical trials or preclinical evidence. METHODS: NKTCL cell line xenograft models (CDX) in immunocompetent, human programmed cell death protein 1 (PD1) knock-in genetically engineered mice were used to investigate the combination effects. Different types and dosages of HDAC inhibitors were investigated. We explored the underlying mechanisms by RNA-sequencing and ChIP-sequencing. Two clinical cases treated with anti-PD1/chidamide were presented. FINDINGS: Anti-PD1/chidamide shows significant tumour rejection in two CDX models. RNA-seq and CHIP-seq revealed that chidamide is synergistic to enhance T-cell chemokine expression, augment the Ifn-γ response, and increase CD8 T-cell infiltration via histone modification. Ifn-γ neutralizing antibody can attenuate the efficacy of combination drugs. However, the anti-PD1/romidepsin failed to augment the Ifn-γ response. The expressions of Ifn-γ related gene set signatures are significantly correlated with tumour rejection in anti-PD1/chidamide. In the clinic, two NKTCL patients treated with the PD1/chidamide show promising efficacy and limited toxicity. INTERPRETATION: Anti-PD1/chidamide enhances T-cell chemokine expression and augments the IFN-γ response in preclinical NKTCL immunocompetent models. IFN-γ signatures may be good response biomarkers for the selection of potentially benefit patients. FUNDING: This study was supported by the Chinese National Major Project for New Drug Innovation (2017ZX09304015) and the Chinese Society of Clinical Oncology Research Fund (Y-BMS2019-026).


Subject(s)
Chemokines , Interferon-gamma , Lymphoma, T-Cell , Animals , Humans , Mice , Antibodies/pharmacology , Antibodies/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemokines/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Lymphoma/drug therapy , Lymphoma, T-Cell/drug therapy , Neoplasm Recurrence, Local/drug therapy , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism
3.
Clin Exp Dermatol ; 47(1): 110-113, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34236726

ABSTRACT

The management of pemphigus vulgaris (PV) is challenging. This study aimed to evaluate the immunomodulating effects of metformin on PV. The study was conducted in two phases: in the first phase, patients received routine first-line treatment (prednisolone plus azathioprine) for 2 months, then in the second phase, metformin was added to this regimen for another 2 months. After addition of metformin to the first-line medications, significant reductions were seen in serum IgG1 (reduced from 534.92 ± 134.83 mg/dL to 481.58 ± 130.46 mg/dL, P < 0.001), IgG4 (51.83 ± 27.26 mg/dL to 44.50 ± 26.05 mg/dL, P < 0.001) and interferon-γ (277.99 ± 108.71 pg/mL to 45.05 ± 17.080 pg/mL, P = 0.03) concentrations. The suppressant effect of metformin was greatest on IgG4 (coefficient of variation 1.28), the dominant subclass of IgG involved in PV. Metformin could have immunomodulating effects on PV with controlling effects on steroid complications.


Subject(s)
Immunoglobulin G/blood , Interferon-gamma/blood , Metformin/therapeutic use , Pemphigus/blood , Pemphigus/drug therapy , Adult , Female , Humans , Immunoglobulin G/drug effects , Interferon-gamma/drug effects , Male , Metformin/pharmacology , Middle Aged , Pemphigus/immunology , Prospective Studies
4.
Cell Mol Life Sci ; 78(24): 8229-8242, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34741187

ABSTRACT

Mitogen-activated protein kinase (MAPK) signalling pathways are crucial for developmental processes, oncogenesis, and inflammation, including the production of proinflammatory cytokines caused by reactive oxygen species and upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. There are no drugs that can effectively prevent excessive inflammatory responses in endothelial cells in the lungs, heart, brain, and kidneys, which are considered the main causes of severe coronavirus disease 2019 (COVID-19). In this work, we demonstrate that human MAPKs, i.e. extracellular signal-regulated kinases 1 and 2 (ERK1/2), are CO2 sensors and CO2 is an efficient anti-inflammatory compound that exerts its effects through inactivating ERK1/2 in cultured endothelial cells when the CO2 concentration is elevated. CO2 is a potent inhibitor of cellular proinflammatory responses caused by H2O2 or the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2. ERK1/2 activated by the combined action of RBD and cytokines crucial for the development of severe COVID-19, i.e. interferon-gamma (IFNγ) and tumour necrosis factor-α (TNFα), are more effectively inactivated by CO2 than by dexamethasone or acetylsalicylic acid in human bronchial epithelial cells. Previously, many preclinical and clinical studies showed that the transient application of 5-8% CO2 is safe and effective in the treatment of many diseases. Therefore, our research indicates that CO2 may be used for the treatment of COVID-19 as well as the modification of hundreds of cellular pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , COVID-19 Drug Treatment , Carbon Dioxide/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , COVID-19/immunology , COVID-19/pathology , Cell Line , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , Inflammation/drug therapy , Interferon-gamma/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Domains/drug effects , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/metabolism , Tumor Necrosis Factor-alpha/drug effects
5.
Pak J Pharm Sci ; 34(3): 971-979, 2021 May.
Article in English | MEDLINE | ID: mdl-34602421

ABSTRACT

Aconitine, the main component in Radix Aconiti Lateralis Preparata, not only exerts the anti-tumor effect on Hepatocellular Carcinoma (HCC) but also damages on immune system. In the present study, Crude Monkshood Polysaccharide (CMP), another one natural composition component originated from the same herbal with aconitine, combined with aconitine to investigate the effects on HCC and immunity in vitro and in vivo. The combination of CMP and aconitine enhanced the ability of the immunocyte to kill the tumor cell in vitro and had an additive effect on anti-HCC in vivo. Aconitine-CMP in combination improved the spleen weights, spleen index, thymus weights, thymus index. Elevated CD4+ T and CD8+ T cells and macrophages in spleen, decreased serum IL-6 level and increased serum IFN-γ and TNF-α levels were observed in mice treated with the combination of aconitine and CMP compare with control group (P<0.05). Our results showed that the combination of aconitine and CMP exerts anti-tumor effect by directly killing tumor cells and enhancing the anti-tumor immune responses, which further implies that chemotherapy drugs combined with Chinese medicine immunopotentiator maybe a feasible and effective strategy for HCC.


Subject(s)
Aconitine/pharmacology , Aconitum , Carcinoma, Hepatocellular/immunology , Cell Proliferation/drug effects , Liver Neoplasms/immunology , Plant Extracts/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , In Vitro Techniques , Interferon-gamma/drug effects , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Liver Neoplasms/metabolism , Lymphocytes/drug effects , Lymphocytes/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Neoplasm Transplantation , Organ Size/drug effects , Polysaccharides/pharmacology , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
Reprod Biol Endocrinol ; 19(1): 128, 2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34429116

ABSTRACT

BACKGROUND: Endometriosis is a serious reproductive and general health consequences. Recombinant human IL-37 (rhIL-37) is an inhibitor of inflammation. METHODS: ELISA assay was performed to detect the concentration of cytokines. Flow cytometry was used to analyze cell proportion. Besides, qRT-PCR and western blotting assay were used to detect the level of gene and protein, respectively. Transwell co-culture system was used for the co-culture of dendritic cells (DCs) and CD4+T cells. RESULTS: Our data showed that rhIL-37 inhibited the development of ectopic lesions in the mice with endometriosis, increased Th1/Th2 ratio and induced DCs maturation. The co-culture system of DCs and CD4+T cells demonstrated that rhIL-37 increased Th1/Th2 cell ratio through promoting DCs maturation. Moreover, the expression of IL-4 in the DCs derived from healthy mice was inhibited by rhIL-37 treatment. rhIL-37 increased Th1/Th2 cell ratio through inhibiting IL-4 in DCs. Subsequently, our results proved that rhIL-37 promoted the maturation of DCs via inhibiting phosphorylation of STAT3. Activation of STAT3 could reverse rhIL-37-induced maturation of DCs. CONCLUSION: Overall, rhIL-37 could protect against endometriosis through increasing the ratio of Th1/Th2 cells via inducing DCs maturation and inhibiting IL-4 expression in the DCs. Furthermore, rhIL-37 induced DCs maturation by inhibiting STAT3 phosphorylation. Our data confirmed the protective effect of rhIL-37 in endometriosis. These data may provide a novel idea for the treatment of the disease.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Dendritic Cells/drug effects , Endometriosis/immunology , Interleukin-1/pharmacology , Th1-Th2 Balance/drug effects , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Endometriosis/metabolism , Endometrium/transplantation , Female , Gene Expression/drug effects , Humans , Interferon-gamma/drug effects , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Phosphorylation , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Recombinant Proteins , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
7.
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
8.
Exp Oncol ; 43(1): 7-14, 2021 03.
Article in English | MEDLINE | ID: mdl-33785718

ABSTRACT

BACKGROUND: Toll-like receptor 4 (TLR4) is known to be involved in carcinogenesis and cancer progression. Changes in TLR4 expression are associated with changes in the expression of key cellular cytokines (transforming growth factor-ß (TGF-ß), tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ)), which affect cancer progression and metastasis. AIM: To study changes in the expression of TLR4, TGF-ß, TNF-α, IFN-γ genes, the level of apoptosis and cell cycle distribution in human invasive urothelial carcinoma T24/83 cells under the treatment with polyphenolic adjuvant compound of fungal origin melanin, cytotoxic drug cisplatin, and combination of both. MATERIALS AND METHODS: T24/83 cells were incubated with cisplatin (0.05 mM), melanin (5 µg/ml), or their combination. The expression level of TLR-4, TGF-ß, INF-γ, TNF-α was evaluated by the real time polymerase chain reaction. The flow cytometry was used to study cell cycle distribution, proliferative activity and level of apoptosis. Morphological analysis of the Т24/83 cells was performed as well. RESULTS: Melanin, cisplatin, and their combination downregulate TLR4 expression (2.67; 1.28; and 2.73-fold decrease, respectively) and TNF-α expression (6.5; 1.4; and 1.7-fold decrease, respectively). Melanin did not affect TGF-ß expression while cisplatin caused 13-fold downregulation of TGF-ß. The combined use of cisplatin and melanin decreased TGF-ß expression by 6.5 times. The upregulation of IFN-γ by melanin, cisplatin, and their combination was demonstrated (4.3; 6.7; and 2-fold increase, respectively). All treatment modalities increased the level of apoptosis in T24/83 cells. Melanin treatment increased significantly the proportion of fibroblast-like cells in T24/83 culture with decreased cell adhesion to the substrate. CONCLUSIONS: Melanin, cisplatin, and combination of both agents affect significantly TLR4, TNF-α, TGF-ß, INF-γ expression, cell cycle distribution and morphology in T24/83 cells suggesting their transition to less aggressive phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Transitional Cell/pathology , Cisplatin/pharmacology , Melanins/pharmacology , Urinary Bladder Neoplasms/pathology , Carcinoma, Transitional Cell/metabolism , Cell Line, Tumor , Humans , Interferon-gamma/drug effects , Toll-Like Receptor 4/drug effects , Transforming Growth Factor beta/drug effects , Tumor Necrosis Factor-alpha/drug effects , Urinary Bladder Neoplasms/metabolism
9.
Int J Neurosci ; 131(6): 591-598, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32250189

ABSTRACT

AIM OF THE STUDY: Glioblastoma multiforme (GBM) is the most severe forms of brain cancer, eventually becoming the leading cause of brain cancer-related death worldwide. Owing to the bleak surgical interventions and resistance to the different treatment regime, GBM is a parlous disease demanding newer therapeutical perspective for its treatment. Toll-like receptors (TLRs) are well-known members of pathogen recognition receptors (PRRs) and have been extensively explored for their therapeutic and prophylactic potential in an array of disease including cancer. Recent trends in drug delivery research has shown shift towards delivering short DNA sequences (CpG DNA) to endosomal TLR9 within immune cells (macrophages, dendritic cells, etc.) for the activation of desired inflammatory response using non-agonistic ß-glucan particles; a well-known ligand for Dectin-1 receptors. Our study is therefore focused to explore the role of nano-encapsulated CpG ODN as critical players in polarizing M2 scavenging to much desired pro-inflammatory type. MATERIALS AND METHODS: The nanoparticles entrapping CpG ODN 1826 were prepared by using a fungal polymer Schizophyllan (SPG). The constructed nanoparticles were characterized and assessed for their efficacy on rat glioblastoma cells (C6). RESULTS: The constructed Schizophyllan (SPG) nanoparticles entrapping CpG ODN 1826 (95.3%) were of 25.49 nm in diameter and thus capable of crossing blood-brain barrier. The rat glioblastoma (C6) cells evaluated for intracellular oxidative burst and cytokine levels pre- and post-incubation with nanoparticles exhibited marked elevation in the expression of intracellular ROS and IFN-γ as well as IL-1ß post treatment. CONCLUSION: The findings indicate towards potentiality of repolarizing the M2 macrophages to much desired M1 phase by inducing higgh levels of oxidative burst and inflammatory cytokines. Consequently, the apoptosis was induced in glioblastoma cells establishing the suitablity of CpG ODN carrying nanoformulations as emerging therapeutic intervention for GBM.


Subject(s)
Adjuvants, Immunologic , Brain Neoplasms/drug therapy , Cytokines/drug effects , Glioblastoma/drug therapy , Lectins, C-Type , Macrophages/drug effects , Nanoparticles , Oligodeoxyribonucleotides , Sizofiran , Toll-Like Receptor 9/agonists , Adjuvants, Immunologic/administration & dosage , Animals , Cell Line, Tumor , Cytokines/metabolism , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Rats , Reactive Oxygen Species/metabolism , Sizofiran/administration & dosage
10.
Int Immunopharmacol ; 88: 106858, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32795895

ABSTRACT

Increasing studies confirm that anti-angiogenesis can increase the effectiveness of immunotherapy. In this study, we found that an angiogenesis inhibitor apatinib enhanced anti-PD-1 therapy for colon cancer in mice via promoting PD-L1 expression. Apatinib treatment upregulated PD-L1 expression in various colon cancer cells both at the mRNA and protein levels. Further, apatinib-treated cancer cells hampered activation and IFN-γ secretion of T cells in the co-culture system, which was reversed by the anti-PD-1 antibody. Based on this, the combination of apatinib with anti-PD-1 on colon cancer growth in mice was examined. The combination treatment showed more significant inhibition on the growth of transplanted tumors in mice than single-drug treatment. Overall, our study here showed the enhancement of anti-PD-1 antitumor efficacy in a syngeneic mouse model (CT-26 cells in Balb/c) by the angiogenesis inhibitor apatinib via upregulating PD-L1 expression as well as angiogenesis inhibition, which may provide a rationale for the combination of apatinib and anti-PD-1 antibody for colorectal cancer treatment in the clinic.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , B7-H1 Antigen/biosynthesis , Colonic Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyridines/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/drug effects , B7-H1 Antigen/genetics , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Female , Granzymes/metabolism , Immunity/drug effects , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Pyridines/therapeutic use , T-Lymphocytes/drug effects , Transplantation, Isogeneic
11.
Parasit Vectors ; 13(1): 343, 2020 Jul 10.
Article in English | MEDLINE | ID: mdl-32650837

ABSTRACT

BACKGROUND: Poultry coccidiosis is a parasitic enteric disease with a highly negative impact on chicken production. In-feed chemoprophylaxis remains the primary method of control, but the increasing ineffectiveness of anticoccidial drugs, and potential future restrictions on their use has encouraged the use of commercial live vaccines. Availability of such formulations is constrained by their production, which relies on the use of live chickens. Several experimental approaches have been taken to explore ways to reduce the complexity and cost of current anticoccidial vaccines including the use of live vectors expressing relevant Eimeria proteins. We and others have shown that vaccination with transgenic Eimeria tenella parasites expressing Eimeria maxima Apical Membrane Antigen-1 or Immune Mapped Protein-1 (EmAMA1 and EmIMP1) partially reduces parasite replication after challenge with a low dose of E. maxima oocysts. In the present study, we have reassessed the efficacy of these experimental vaccines using commercial birds reared at high stocking densities and challenged with both low and high doses of E. maxima to evaluate how well they protect chickens against the negative impacts of disease on production parameters. METHODS: Populations of E. tenella parasites expressing EmAMA1 and EmIMP1 were obtained by nucleofection and propagated in chickens. Cobb500 broilers were immunised with increasing doses of transgenic oocysts and challenged two weeks later with E. maxima to quantify the effect of vaccination on parasite replication, local IFN-γ and IL-10 responses (300 oocysts), as well as impacts on intestinal lesions and body weight gain (10,000 oocysts). RESULTS: Vaccination of chickens with E. tenella expressing EmAMA1, or admixtures of E. tenella expressing EmAMA1 or EmIMP1, was safe and induced partial protection against challenge as measured by E. maxima replication and severity of pathology. Higher levels of protection were observed when both antigens were delivered and was associated with a partial modification of local immune responses against E. maxima, which we hypothesise resulted in more rapid immune recognition of the challenge parasites. CONCLUSIONS: This study offers prospects for future development of multivalent anticoccidial vaccines for commercial chickens. Efforts should now be focused on the discovery of additional antigens for incorporation into such vaccines.


Subject(s)
Chickens/parasitology , Coccidiosis/veterinary , Eimeria tenella , Protozoan Vaccines , Animals , Antigens, Protozoan/immunology , Body Weight/drug effects , Chickens/immunology , Coccidiosis/prevention & control , Coccidiosis/therapy , Eimeria/drug effects , Eimeria/growth & development , Eimeria/immunology , Eimeria tenella/drug effects , Eimeria tenella/growth & development , Eimeria tenella/immunology , Genes, Protozoan/immunology , Interferon-gamma/drug effects , Interleukin-10/metabolism , Poultry Diseases/parasitology , Poultry Diseases/prevention & control , Protozoan Vaccines/biosynthesis , Protozoan Vaccines/therapeutic use , Transfection , Transgenes/immunology , Vaccination/methods , Vaccination/veterinary , Vaccines, Attenuated/biosynthesis , Vaccines, Attenuated/therapeutic use
12.
Chem Commun (Camb) ; 56(53): 7273-7276, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32478368

ABSTRACT

Nanoparticle-cell-nanoparticle communication by stigmergy was demonstrated using two capped nanodevices. The first community of nanoparticles (i.e.S(RA)IFN) is loaded with 9-cis-retinoic acid and capped with interferon-γ, whereas the second community of nanoparticles (i.e.S(sulf)PIC) is loaded with sulforhodamine B and capped with poly(I:C). The uptake of S(RA)IFN by SK-BR-3 breast cancer cells enhanced the expression of TLR3 receptor facilitating the subsequent uptake of S(sulf)PIC and cell killing.


Subject(s)
Antineoplastic Agents/metabolism , Cell Communication/drug effects , Interferon Inducers/metabolism , Nanoparticles/chemistry , Poly I-C/metabolism , Alitretinoin/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Interferon Inducers/chemistry , Interferon-gamma/drug effects , Nanoparticles/metabolism , Poly I-C/chemistry , Rhodamines/chemistry , Toll-Like Receptor 3/genetics
13.
Folia Med (Plovdiv) ; 62(1): 37-45, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32337895

ABSTRACT

BACKGROUND: The purpose of the present study was to produce a pcDNA3.1(+)-ureA recombinant vector and evaluate the capacity of this vector to stimulate the immune response against H. pylori infection in infused BALB/c mice. MATERIALS AND METHODS: The pcDNA3.1(+)-ureA construct was prepared and transformed into E. coli, successfully. The animals we used in the study were allotted into three groups for infusion of 1) recombinant plasmid, 2) pcDNA3.1(+)-ureA + nanoparticles, and 3) pcDNA3.1(+). Blood and tissue specimens from each group of mice were collected at days 15, 30, and 45 after the last infusion and the expression levels of cytokines such as TGF-ß1, IL-4, and IFNγ genes comparing to GAPDH as well as the expression of ureA in the mice's thigh muscle were evaluated. RESULTS: The genes expression analysis showed that the IL4 expression significantly decreased (p<0.001) but IFNγ and TGF-ß1 expression increased in the blood of infused mice (p<0.001). Also, the urea expression level in pcDNA3.1(+)-urea and pcDNA3.1(+)-ureA+ nanoparticle 15, 30, and 45 days after the last infusion was significantly different (p<0.001) and its expressions at days 15 and 30 were significantly different (p<0.001), but 45 days after the last infusion it was not significantly different (p>0.05). CONCLUSION: The pcDNA3.1(+)-ureA recombinant vector with or without chitosan nanoparticles can stimulate the immune response in animal models against H. pylori infection. Also, after combining the recombinant vector with nanoparticles we observed a better immune response was observed. In future studies this recombinant construct can be used as a biomarker and therapeutic approaches in eukaryotic systems.


Subject(s)
Bacterial Proteins/genetics , Cytokines/drug effects , Helicobacter pylori/genetics , Immunity/drug effects , Urease/genetics , Vaccines, DNA/pharmacology , Animals , Chitosan , Cytokines/genetics , Female , Helicobacter Infections , Interferon-gamma/drug effects , Interferon-gamma/genetics , Interleukin-4/genetics , Mice , Mice, Inbred BALB C , Nanoparticles , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/genetics
14.
EMBO J ; 39(10): e103758, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32293748

ABSTRACT

The protozoan parasite Toxoplasma gondii lives inside a vacuole in the host cytosol where it is protected from host cytoplasmic innate immune responses. However, IFNγ-dependent cell-autonomous immunity can destroy the vacuole and the parasite inside. Toxoplasma strain differences in susceptibility to human IFNγ exist, but the Toxoplasma effector(s) that determine these differences are unknown. We show that in human primary fibroblasts, the polymorphic Toxoplasma-secreted effector GRA15 mediates the recruitment of ubiquitin ligases, including TRAF2 and TRAF6, to the vacuole membrane, which enhances recruitment of ubiquitin receptors (p62/NDP52) and ubiquitin-like molecules (LC3B, GABARAP). This ultimately leads to lysosomal degradation of the vacuole. In murine fibroblasts, GRA15-mediated TRAF6 recruitment mediates the recruitment of immunity-related GTPases and destruction of the vacuole. Thus, we have identified how the Toxoplasma effector GRA15 affects cell-autonomous immunity in human and murine cells.


Subject(s)
Foreskin/parasitology , Interferon-gamma/pharmacology , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/parasitology , Foreskin/cytology , Foreskin/metabolism , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Signal Transduction , Toxoplasma/metabolism , Vacuoles/metabolism
15.
Cancer Med ; 9(10): 3584-3591, 2020 05.
Article in English | MEDLINE | ID: mdl-32212317

ABSTRACT

Ovarian cancer (OC) is the fifth-leading cause of cancer-related death in women with a pathogenesis involving activation of regulatory T cells (Tregs). The T-cell immunoglobulin and ITIM domain (TIGIT) is a well-known immune checkpoint molecule that inhibits T-cell responses. However, the role of TIGIT in OC is not comprehensively understood. In this study, we revealed crucial functions of TIGIT in the development and progression of OC. ID8 cells were used to establish a murine OC model. TIGIT expression was increased in immune cells of OC mice, particularly in CD4+ Tregs. Anti-TIGIT monoclonal antibodies (mAb) were used to block the function of TIGIT in OC mice, and we found that the anti-TIGIT treatment reduced the proportion of CD4+ Tregs, but did not affect CD4+ and CD8+ T cells or natural killer cells. Splenic CD4+ Tregs from OC mice were isolated after the anti-TIGIT treatment, and their functioning was examined. Inhibition of TIGIT lowered the degree of immunosuppression induced by CD4+ Tregs. A survival curve suggested that anti-TIGIT treatment can improve the survival rate of OC in mice. We conclude that TIGIT enhanced CD4+ Tregs response and mediated immunosuppression in the OC model. Our data suggest that inhibition of TIGIT is a potential therapeutic target in OC patients.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , Ovarian Neoplasms/metabolism , Receptors, Immunologic/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Ovarian Epithelial/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Interferon-gamma/drug effects , Interferon-gamma/immunology , Interleukin-4/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Ovarian Neoplasms/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Escape/immunology
16.
World Neurosurg ; 138: 758-763, 2020 06.
Article in English | MEDLINE | ID: mdl-32004735

ABSTRACT

OBJECTIVE: We sought to investigate the effects and mechanism of lead and a high-fat diet on cognitive function and the central nervous system in mice. METHODS: Eighty-four healthy male mice were randomly divided into a control group (n = 21) (fed with common diet and free drinking), a lead exposure group (n = 21) (fed with common diet and 300 mg/L lead acetate solution), a high-fat group (n = 21) (fed with high-fat diet and free drinking), and a lead + high-fat group (n = 21) (fed with high-fat diet and 300 mg/L lead acetate solution). In 10 weeks after lead exposure, the mice of all groups were tested for the cognition, learning and memory abilities, body weight, serum triglyceride (TG), low-density lipoprotein, and high-density lipoprotein, as well as for the contents of lead, interleukin 6 (IL-6), interleukin 17 (IL-17), interferon γ, advanced glycation end products (AGEs), glutathione S-transferase (GSH-ST), and hydrogen peroxide in the brain tissues. RESULTS: Compared with the control group and the lead-exposed group, the body weights of mice in the high-fat group and the lead + high-fat group increased significantly from the sixth week of the experiment, of which the difference was statistically significant (P < 0.05). Compared with the control group and the high-fat group, the lead content in brain tissue of the lead exposure group and the lead + high-fat group increased significantly, of which the difference was statistically significant (P < 0.05). Compared with the control group, the escape latent period, triglyceride, low-density lipoprotein, IL-6, IL-17, interferon γ, and AGEs of the remaining 3 groups increased significantly, but the recognition index, passing platform times, high-density lipoprotein, and GSH-ST significantly decreased (P < 0.05); the second and third escape latent periods, IL-6, IL-17, and AGEs of lead + high-fat group, were obviously higher than the remaining 3 groups, but the passing platform times were obviously lower than the remaining 3 groups, of which the difference was statistically significant. The content of hydrogen peroxide in brain tissues had no difference among groups (P > 0.05). CONCLUSIONS: The lead and high-fat diet resulted in lipid metabolism disorders and impaired the cognitive function and central nervous system by promoting the secretion of inflammatory factors in glial cells, inducing the inflammatory reaction of brain tissue, inhibiting GSH-ST expression, and increasing AGEs content.


Subject(s)
Brain/drug effects , Cognition/drug effects , Diet, High-Fat , Lead Poisoning/psychology , Lead/toxicity , Animals , Brain/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Glycation End Products, Advanced/drug effects , Glycation End Products, Advanced/metabolism , Hydrogen Peroxide/metabolism , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Lead Poisoning/metabolism , Lipoproteins, HDL/drug effects , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/drug effects , Lipoproteins, LDL/metabolism , Male , Mice , Random Allocation , Triglycerides/metabolism
17.
Respir Physiol Neurobiol ; 276: 103412, 2020 05.
Article in English | MEDLINE | ID: mdl-32044448

ABSTRACT

Allergic asthma is a chronic inflammatory disease characterized by airflow obstruction, airway hyperresponsiveness (AHR), airway inflammation, and mucus overproduction. Cordyceps polysaccharide (CPS) is one of the main bioactive compounds of Cordyceps militarisis, a traditional Chinese medicine. In this study, we established a mouse model of asthma using ovalbumin (OVA) challenge and evaluated the potential regulatory effect of CPS (25, 50, and 100 mg/kg) on asthmatic mice. These results showed that the asthmatic mice treated with CPS suppressed the secretion of eotaxin, IL-4, IL-5, IL-13, and IFN-γ in the blood and bronchoalveolar lavage fluid (BALF), and decreased serum IgE levels compared to the vehicle-treated mice. CPS also alleviated inflammatory cell infiltration, goblet cell hyperplasia, and the increases of inflammatory cells in the mouse model of asthma. In addition, OVA-induced AHR was inhibited by CPS treatment. Further analyses of protein expression revealed that CPS inhibited the activation of transforming growth factor ß1 (TGF-ß1)/Smad pathway in mice with asthma. These findings indicated that CPS might serve as a potential therapeutic agent for the management of allergic asthma.


Subject(s)
Asthma/metabolism , Cordyceps , Fungal Polysaccharides/pharmacology , Lung/drug effects , Smad2 Protein/drug effects , Smad3 Protein/drug effects , Transforming Growth Factor beta1/drug effects , Animals , Asthma/chemically induced , Asthma/physiopathology , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Interleukin-5/metabolism , Lung/metabolism , Lung/physiopathology , Medicine, Chinese Traditional , Mice , Ovalbumin , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/physiopathology , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
18.
Clin Microbiol Infect ; 26(11): 1555.e1-1555.e7, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32062048

ABSTRACT

OBJECTIVES: Using QuantiFERON-TB Gold In-Tube (QFT-GIT) for monitoring tuberculosis (TB) and latent TB infection treatment effect is controversial. The present study aimed to evaluate the dynamic changes of interferon gamma (IFN-γ) levels along with latent TB infection treatment via a randomized controlled study. METHODS: A total of 910 participants treated with 8 weeks of once-weekly rifapentine plus isoniazid (group A), 890 treated with 6 weeks of twice-weekly rifapentine plus isoniazid (group B) and 818 untreated controls (group C) were followed for 2 years to track active TB development. QFT-GIT tests were repeated three times for all groups: before treatment (T0), at completion of treatment (T1) and 3 months after completion of treatment (T2). RESULTS: Similar rates of persistent QFT-GIT reversion were observed in groups A (19.0%, 173/910), B (18.5%, 165/890) and C (20.7%, 169/818) (p 0.512). The dynamic changes of IFN-γ levels were not statistically significant among the three groups. In treated participants, individuals with higher baseline IFN-γ levels showed increased TB occurrence (1.0%, 9/896) compared to those with lower baseline levels (0.2%, 2/904) (p 0.037). A similar but statistically insignificant trend was also observed in untreated controls (1.8% (7/400) vs. 0.5% (2/418), p 0.100). When TB cases were matched with non-TB cases on baseline IFN-γ levels, no significant differences were found with respect to the dynamic changes in IFN-γ levels with time, regardless of whether they received treatment. CONCLUSIONS: QFT-GIT reversion or decreased IFN-γ levels should not be used for monitoring host response to latent TB infection treatment.


Subject(s)
Antitubercular Agents/administration & dosage , Interferon-gamma/metabolism , Isoniazid/administration & dosage , Latent Tuberculosis/drug therapy , Rifampin/analogs & derivatives , Antitubercular Agents/pharmacology , Biomarkers/metabolism , China , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Interferon-gamma/drug effects , Interferon-gamma Release Tests , Isoniazid/pharmacology , Latent Tuberculosis/immunology , Male , Rifampin/administration & dosage , Rifampin/pharmacology , Treatment Outcome
19.
Clin Exp Allergy ; 50(1): 82-94, 2020 01.
Article in English | MEDLINE | ID: mdl-31579973

ABSTRACT

BACKGROUND: Acute exacerbations of asthma represent a major burden of disease and are often caused by respiratory infections. Viral infections are recognized as significant triggers of exacerbations; however, less is understood about the how microbial bioproducts such as the endotoxin (lipopolysaccharide (LPS)) trigger episodes. Indeed, increased levels of LPS have been linked to asthma onset, severity and steroid resistance. OBJECTIVE: The goal of this study was to identify mechanisms underlying bacterial-induced exacerbations by employing LPS as a surrogate for infection. METHODS: We developed a mouse model of LPS-induced exacerbation on the background of pre-existing type-2 allergic airway disease (AAD). RESULTS: LPS-induced exacerbation was characterized by steroid-resistant airway hyperresponsiveness (AHR) and an exaggerated inflammatory response distinguished by increased numbers of infiltrating neutrophils/macrophages and elevated production of lung inflammatory cytokines, including TNFα, IFNγ, IL-27 and MCP-1. Expression of the type-2 associated inflammatory factors such as IL-5 and IL-13 were elevated in AAD but not altered by LPS exposure. Furthermore, AHR and airway inflammation were no longer suppressed by corticosteroid (dexamethasone) treatment after LPS exposure. Depletion of pulmonary macrophages by administration of 2-chloroadenosine into the lungs suppressed AHR and reduced IL-13, TNFα and IFNγ expression. Blocking IL-13 function, through either IL-13-deficiency or administration of specific blocking antibodies, also suppressed AHR and airway inflammation. CONCLUSIONS & CLINICAL RELEVANCE: We present evidence that IL-13 and innate immune pathways (in particular pulmonary macrophages) contribute to LPS-induced exacerbation of pre-existing AAD and provide insight into the complex molecular processes potentially underlying microbial-induced exacerbations.


Subject(s)
Asthma/immunology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Interleukin-13/immunology , Lipopolysaccharides/pharmacology , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Respiratory Hypersensitivity/immunology , Airway Resistance/drug effects , Animals , Bacterial Infections , Bronchoalveolar Lavage Fluid/cytology , Chemokine CCL2 , Cytokines/drug effects , Cytokines/immunology , Disease Models, Animal , Disease Progression , Drug Resistance , Interferon-gamma/drug effects , Interferon-gamma/immunology , Interleukins/immunology , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Mice , Mucin 5AC/drug effects , Mucin 5AC/metabolism , Ovalbumin , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/immunology
20.
Inflamm Bowel Dis ; 26(4): 534-545, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31634391

ABSTRACT

BACKGROUND: Intestinal epithelial apical membrane Cl-/HCO3- exchanger DRA (downregulated in adenoma, SLC26A3) has emerged as an important therapeutic target for diarrhea, emphasizing the potential therapeutic role of agents that upregulate DRA. All-trans retinoic acid (ATRA), a key vitamin A metabolite, was earlier shown by us to stimulate DRA expression in intestinal epithelial cells. However, its role in modulating DRA in gut inflammation has not been investigated. AIMS: Our aim was to analyze the efficacy of ATRA in counteracting inflammation-induced decrease in DRA in vitro and in vivo. METHODS: Interferon-γ (IFN-γ)-treated Caco-2 cells and dextran sulfate sodium (DSS)-treated C57BL/6J mice served as in vitro and in vivo models of gut inflammation, respectively. The effect of ATRA on IFN-γ-mediated inhibition of DRA function, expression, and promoter activity were elucidated. In the DSS colitis model, diarrheal phenotype, cytokine response, in vivo imaging, myeloperoxidase activity, and DRA expression were measured in the distal colon. RESULTS: All-trans retinoic acid (10 µM, 24 h) abrogated IFN-γ (30 ng/mL, 24 h)-induced decrease in DRA function, expression, and promoter activity in Caco-2 cells. All-trans retinoic acid altered IFN-γ signaling via blocking IFN-γ-induced tyrosine phosphorylation of STAT-1. All-trans retinoic acid cotreatment (1 mg/kg BW, i.p. daily) of DSS-treated mice (3% in drinking water for 7 days) alleviated colitis-associated weight loss, diarrheal phenotype, and induction of IL-1ß and CXCL1 and a decrease in DRA mRNA and protein levels in the colon. CONCLUSION: Our data showing upregulation of DRA under normal and inflammatory conditions by ATRA demonstrate a novel role of this micronutrient in alleviating IBD-associated diarrhea.


Subject(s)
Antiporters/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Colitis/drug therapy , Intestinal Mucosa/drug effects , Sulfate Transporters/metabolism , Tretinoin/pharmacology , Animals , Antiporters/genetics , Caco-2 Cells , Chloride-Bicarbonate Antiporters/genetics , Colon/metabolism , Dextran Sulfate/toxicity , Diarrhea/drug therapy , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Inflammation/metabolism , Interferon-gamma/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Sulfate Transporters/genetics , Up-Regulation , Weight Loss/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL