Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
2.
Int Immunopharmacol ; 126: 111300, 2024 Jan 05.
Article En | MEDLINE | ID: mdl-38016346

Encapsulating peritoneal sclerosis (EPS) is a severe complication of peritoneal dialysis (PD). Surgery is a therapeutic strategy for the treatment of complete intestinal obstruction. However, complete intestinal obstruction in long-term PD results in high mortality and morbidity rates after surgery. Immunopathogenesis participates in EPS formation: CD8, Th1, and Th17 cell numbers increased during the formation of EPS. The anti-inflammatory and immunomodulatory effects of melatonin may have beneficial effects on this EPS. In the present study, we determined that melatonin treatment significantly decreases the Th1 and Th17 cell populations in mice with EPS, decreases the production of IL-1ß, TNF-α, IL-6, and IFN-γ, and increases the production of IL-10. The suppression of Th1 and Th17 cell differentiation by melatonin occurs through the inhibition of dendritic cell (DC) activation by affecting the initiation of the NF-κB signaling pathway in DCs. Our study suggests that melatonin has preventive potential against the formation of EPS in patients with PD.


Intestinal Obstruction , Melatonin , Peritoneal Fibrosis , Humans , Animals , Mice , Peritoneal Fibrosis/etiology , NF-kappa B/metabolism , Melatonin/pharmacology , Melatonin/therapeutic use , Cell Differentiation , Signal Transduction , Dendritic Cells/metabolism , Intestinal Obstruction/complications , Intestinal Obstruction/pathology
3.
Biomedicines ; 11(10)2023 Sep 29.
Article En | MEDLINE | ID: mdl-37893043

The dysregulated expression of cyclin genes can lead to the uncontrolled proliferation of cancer cells. Histone demethylase Jumonji-C domain-containing protein 5 (KDM8, JMJD5) and cyclin A1 (CCNA1) are pivotal in cell cycle progression. A promising candidate for augmenting cancer treatment is Allyl isothiocyanate (AITC), a natural dietary chemotherapeutic and epigenetic modulator. This study aimed to investigate AITC's impact on the KDM8/CCNA1 axis to elucidate its role in oral squamous cell carcinoma (OSCC) tumorigenesis. The expression of KDM8 and CCNA1 was assessed using a tissue microarray (TMA) immunohistochemistry (IHC) assay. In vitro experiments with OSCC cell lines and in vivo experiments with patient-derived tumor xenograft (PDTX) and SAS subcutaneous xenograft tumor models were conducted to explore AITC's effects on their expression and cell proliferation. The results showed elevated KDM8 and CCNA1 levels in the OSCC patient samples. AITC exhibited inhibitory effects on OSCC tumor growth in vitro and in vivo. Additionally, AITC downregulated KDM8 and CCNA1 expression while inducing histone H3K36me2 expression in oral cancer cells. These findings underscore AITC's remarkable anticancer properties against oral cancer, highlighting its potential as a therapeutic option for oral cancer treatment by disrupting the cell cycle by targeting the KDM8/CCNA1 axis.

4.
Cell Death Dis ; 14(6): 369, 2023 06 21.
Article En | MEDLINE | ID: mdl-37344492

Acute lung injury (ALI) is characterised by severe pulmonary inflammation, alveolar-capillary barrier disruption, and pulmonary oedema. Therefore, establishing effective therapeutic targets for ALI prevention is crucial. The present study reports a novel function of RNF128 in regulating LPS-induced ALI. Severe lung damage and increased immune cell infiltration were detected in RNF128-deficient mice. In vitro experiments revealed that RNF128 inhibits neutrophil activation by binding to myeloperoxidase (MPO) and reducing its levels and activity. Moreover, RNF128 regulates alveolar macrophage activation and neutrophil infiltration by interacting with TLR4, targeting it for degradation, and inhibiting NF-κB activation, hence decreasing pro-inflammatory cytokines. Our results demonstrate for the first time that RNF128 is a negative regulator of MPO and TLR4 in neutrophils and alveolar macrophages, respectively. However, AAV9-mediated RNF128 overexpression alleviated lung tissue damage and reduced inflammatory cell infiltration. Thus, RNF128 is a promising therapeutic candidate for pharmacological interventions in ALI.


Acute Lung Injury , NF-kappa B , Ubiquitin-Protein Ligases , Animals , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/prevention & control , Lipopolysaccharides/pharmacology , Lung/metabolism , Neutrophil Infiltration , NF-kappa B/metabolism , Peroxidase/metabolism , Toll-Like Receptor 4/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Int J Mol Sci ; 24(8)2023 Apr 19.
Article En | MEDLINE | ID: mdl-37108686

Rheumatoid arthritis (RA) is an autoimmune disease with chronic inflammation, bone erosion, and joint deformation. Synovial tissue in RA patients is full of proinflammatory cytokines and infiltrated immune cells, such as T help (Th) 9, Th17, macrophages, and osteoclasts. Recent reports emphasized a new member of the interleukin (IL)-10 family, IL-26, an inducer of IL-17A that is overexpressed in RA patients. Our previous works found that IL-26 inhibits osteoclastogenesis and conducts monocyte differentiation toward M1 macrophages. In this study, we aimed to clarify the effect of IL-26 on macrophages linking to Th9 and Th17 in IL-9 and IL-17 expression and downstream signal transduction. Murine and human macrophage cell lines and primary culture cells were used and stimulated by IL26. Cytokines expressions were evaluated by flow cytometry. Signal transduction and transcription factors expression were detected by Western blot and real time-PCR. Our results show that IL-26 and IL-9 colocalized in macrophage in RA synovium. IL-26 directly induces macrophage inflammatory cytokines IL-9 and IL-17A expression. IL-26 increases the IL-9 and IL-17A upstream mechanisms IRF4 and RelB expression. Moreover, the AKT-FoxO1 pathway is also activated by IL-26 in IL-9 and IL-17A expressing macrophage. Blockage of AKT phosphorylation enhances IL-26 stimulating IL-9-producing macrophage cells. In conclusion, our results support that IL-26 promotes IL-9- and IL-17-expressing macrophage and might initiate IL-9- and IL-17-related adaptive immunity in rheumatoid arthritis. Targeting IL-26 may a potential therapeutic strategy for rheumatoid arthritis or other IL-9 plus IL-17 dominant diseases.


Arthritis, Rheumatoid , Interleukin-17 , Animals , Humans , Mice , Arthritis, Rheumatoid/metabolism , Cytokines/metabolism , Interleukin-17/genetics , Interleukin-17/pharmacology , Interleukin-17/metabolism , Interleukin-9/metabolism , Macrophages/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Th17 Cells , Interleukins/pharmacology
6.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article En | MEDLINE | ID: mdl-35163121

Type 1 diabetes (T1D) is caused by the destruction of ß cells in pancreatic islets by autoimmune T cells. Islet transplantation has been established as an effective treatment for T1D. However, the survival of islet grafts is often disrupted by recurrent autoimmunity. Alpha-lipoic acid (ALA) has been reported to have immunomodulatory effects and, therefore, may have therapeutic potential in the treatment of T1D. In this study, we investigated the therapeutic potential of ALA in autoimmunity inhibition. We treated non-obese diabetic (NOD) mice with spontaneous diabetes and islet-transplantation mice with ALA. The onset of diabetes was decreased and survival of the islet grafts was extended. The populations of Th1 cells decreased, and regulatory T cells (Tregs) increased in ALA-treated mice. The in vitro Treg differentiation was significantly increased by treatment with ALA. The adoptive transfer of ALA-differentiated Tregs into NOD recipients improved the outcome of the islet grafts. Our results showed that in vivo ALA treatment suppressed spontaneous diabetes and autoimmune recurrence in NOD mice by inhibiting the Th1 immune response and inducing the differentiation of Tregs. Our study also demonstrated the therapeutic potential of ALA in Treg-based cell therapies and islet transplantation used in the treatment of T1D.


Autoimmunity , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/prevention & control , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , T-Lymphocytes, Regulatory/immunology , Thioctic Acid/pharmacology , Animals , Antioxidants/pharmacology , Cell Differentiation , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Female , Graft Survival , Mice , Mice, Inbred NOD , Th1 Cells
7.
Pharmaceuticals (Basel) ; 14(5)2021 May 17.
Article En | MEDLINE | ID: mdl-34067829

Type 1 diabetes mellitus (T1D) results from the destruction of insulin-producing ß cells in the islet of the pancreas by lymphocytes. Non-obese diabetic (NOD) mouse is an animal model frequently used for this disease. It has been considered that T1D is a T cell-mediated autoimmune disease. Both CD4+ and CD8+ T cells are highly responsible for the destruction of ß cells within the pancreatic islets of Langerhans. Previous studies have revealed that regulatory T (Treg) cells play a critical role in the homeostasis of the immune system as well as immune tolerance to autoantigens, thereby preventing autoimmunity. Valproic acid (VPA), a branched short-chain fatty acid, is widely used as an antiepileptic drug and a mood stabilizer. Previous reports have demonstrated that VPA treatment decreases the incidence and severity of collagen-induced arthritis and experimental autoimmune neuritis by increasing the population of Treg cells in these mouse disease models. Given the effect of VPA in the induction of Treg cells' population, we evaluated the therapeutic potential and the protective mechanism of VPA treatment in the suppression of graft autoimmune rejection and immune recurrence in syngeneic or allogenic islet transplantation mouse models. In our study, we found that the treatment of VPA increased the expression of forkhead box P3 (FOXP3), which is a critical transcription factor that controls Treg cells' development and function. Our data revealed that 400 mg/kg VPA treatment in recipients effectively prolonged the survival of syngeneic and allogenic islet grafts. The percentage of Treg cells in splenocytes increased in VPA-treated recipients. We also proved that adoptive transfer of VPA-induced Tregs to the transplanted recipients effectively prolonged the survival of islet grafts. The results of this study provide evidence of the therapeutic potential and the underlying mechanism of VPA treatment in syngeneic islet transplantation for T1D. It also provides experimental evidence for cell therapy by adoptive transferring of in vitro VPA-induced Tregs for the suppression of autoimmune recurrence.

8.
Front Immunol ; 12: 638381, 2021.
Article En | MEDLINE | ID: mdl-33868265

While oxidative stress has been linked to multiple sclerosis (MS), the role of superoxide-producing phagocyte NADPH oxidase (Nox2) in central nervous system (CNS) pathogenesis remains unclear. This study investigates the impact of Nox2 gene ablation on pro- and anti-inflammatory cytokine and chemokine production in a mouse experimental autoimmune encephalomyelitis (EAE) model. Nox2 deficiency attenuates EAE-induced neural damage and reduces disease severity, pathogenic immune cells infiltration, demyelination, and oxidative stress in the CNS. The number of autoreactive T cells, myeloid cells, and activated microglia, as well as the production of cytokines and chemokines, including GM-CSF, IFNγ, TNFα, IL-6, IL-10, IL-17A, CCL2, CCL5, and CXCL10, were much lower in the Nox2-/- CNS tissues but remained unaltered in the peripheral lymphoid organs. RNA-seq profiling of microglial transcriptome identified a panel of Nox2 dependent proinflammatory genes: Pf4, Tnfrsf9, Tnfsf12, Tnfsf13, Ccl7, Cxcl3, and Cxcl9. Furthermore, gene ontology and pathway enrichment analyses revealed that microglial Nox2 plays a regulatory role in multiple pathways known to be important for MS/EAE pathogenesis, including STAT3, glutathione, leukotriene biosynthesis, IL-8, HMGB1, NRF2, systemic lupus erythematosus in B cells, and T cell exhaustion signaling. Taken together, our results provide new insights into the critical functions performed by microglial Nox2 during the EAE pathogenesis, suggesting that Nox2 inhibition may represent an important therapeutic target for MS.


Encephalomyelitis, Autoimmune, Experimental/metabolism , Microglia/metabolism , NADPH Oxidase 2/metabolism , Oxidative Stress/physiology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , NADPH Oxidase 2/immunology
9.
Biomed Pharmacother ; 133: 111057, 2021 Jan.
Article En | MEDLINE | ID: mdl-33378962

Biological and prognostic roles of programmed death ligand 1 (PD-L1) remain unclear in oral squamous cell carcinoma (OSCC). Moreover, the pivotal role of tumor microenvironmental interferon-gamma (IFN-γ) in host responses to malignant cells, oral cancer growth, and PD-L1 expression has not been adequately studied. Thus, PD-L1 expression in 130 OSCC samples was analyzed using immunohistochemistry, which was found significantly overexpressed at the tumor site (P < .01). We further analyzed the effects of IFN-γ on OSCC cell proliferation using enzyme-linked immunosorbent assays and found that IFN-γ drives PD-L1 expression in OSCC cells in a dose-dependent manner. Triptolide (TPL), a bioactive compound isolated from Tripterygium wilfordii, exhibits anti-inflammatory and antitumor activities. To investigate whether the antitumor effect of TPL involves the suppression of PD-L1 expression, we treated OSCC cells in vitro and a patient-derived tumor xenograft (PDTX) model with TPL. TPL suppressed PD-L1 expression in the PDTX model, inhibiting tumor growth, and in OSCC cells in an IFN-γ-modulated microenvironment. We concluded that TPL inhibits tumor growth in oral cancer and downregulates PD-L1 expression in oral cancer cells in vitro. Our results provide evidence for the clinical development of PD-L1-targeted therapy for OSCC.


B7-H1 Antigen/metabolism , Cell Proliferation/drug effects , Diterpenes/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Interferon-gamma/pharmacology , Mouth Neoplasms/drug therapy , Phenanthrenes/pharmacology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Animals , B7-H1 Antigen/genetics , Cell Line, Tumor , Down-Regulation , Epoxy Compounds/pharmacology , Female , Humans , Janus Kinase 2/metabolism , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , STAT1 Transcription Factor/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Burden , Xenograft Model Antitumor Assays
10.
Ecotoxicol Environ Saf ; 202: 110920, 2020 Oct 01.
Article En | MEDLINE | ID: mdl-32800255

Advanced glycation end products (AGE) and the receptor for AGE (RAGE) have been found to be pivotal biomarkers to predict the risk of inflammation and oxidative stress. Limited evidence focuses on the influence of occupational exposure to polycyclic aromatic hydrocarbon (PAH) and metal fumes on AGE and RAGE in shipyard welders. Our aim was to determine the relationships among PAH, metal exposure, and inflammatory biomarkers. From September 1 to December 31, 2017, 53 welding workers (exposed group) and 29 office workers (control group) were enrolled in the study. Comprehensive workups included demographic characteristics, laboratory data, AGE, RAGE, Interleukin-6 (IL-6), tumor necrosis factor-α, PAH, and urinary metal concentrations. RAGE levels were measured by flow cytometric analysis. Urinary 1-hydroxypyrene (1-OHP) was used as a biomarker of exposure to PAH. Several metals were elevated in the personal fine particulate matter (PM2.5) samples, including Mn, Fe, V, Co, Zn, and Cu. The exposed group had significantly higher exposure to PM2.5 (p = 0.015), RAGE (p = 0.020), IL-6 (p = 0.008) than the control group. After adjusting for pertinent variables, there was still a significant and positive association between Ni level and AGE (ß = 0.101; 95% CI, 0.031-0.172). Significant relationship between Cr and Cd levels and RAGE was observed (ß = 0.173; 95% CI, 0.017-0.329; ß = 0.084; 95% CI, 0.011-0.157, respectively). Participants with elevated 1-OHP level had higher odds of high RAGE level in the model 1 (OR = 3.466, 95% CI, 1.053-11.412) and model 2 (OR = 3.454, 95% CI, 1.034-11.536). The RAGE expression of participants was significantly associated with IL-6 levels in the fully adjusted model (ß = 0.294; 95% CI, 0.083-0.732). Our findings highlighted that urinary metal levels and PAH were associated with increased AGE and RAGE formation in shipyard workers. Elevated serum RAGE might induce the production of proinflammatory cytokines and trigger ensuing inflammatory cascades.


Air Pollutants, Occupational/analysis , Metals/analysis , Occupational Exposure/analysis , Polycyclic Aromatic Hydrocarbons/analysis , Receptor for Advanced Glycation End Products/metabolism , Air Pollutants, Occupational/toxicity , Biomarkers/urine , Gases/analysis , Glycation End Products, Advanced/blood , Humans , Inflammation , Male , Metals/toxicity , Oxidative Stress , Particulate Matter/analysis , Polycyclic Aromatic Hydrocarbons/toxicity , Tumor Necrosis Factor-alpha , Welding
11.
PLoS One ; 15(7): e0236101, 2020.
Article En | MEDLINE | ID: mdl-32678829

Dysregulation of histone demethylase Jumonji-C domain-containing protein 5 (JMJD5) has been identified as a great effect on tumorigenesis. Silibinin is a commonly used anti-hepatotoxic drug and exhibits anticancer effect in various cancers. However, the antitumor mechanism between silibinin and JMJD5 in oral squamous cell carcinoma (OSCC) remains unclear. In this study, the clinical significance of JMJD5 on OSCC patients was assessed through tissue microarray. Furthermore, mice bearing patient-derived tumor xenografts (PDTXs) and tongue cancer cell lines were treated with silibinin and evaluated for tumor growth and JMJD5 expression. High expression of JMJD5 in oral cancer was significantly associated with tumor size (P = 0.0241), cervical node metastasis (P = 0.0001) and clinical stage (P = 0.0002), was associated with worse survival rate compared with that of the total cohort (P = 0.0002). Collectively the data indicate that JMJD5 expression may be suitable for detection of unfavorable prognosis in OSCC patients, based in part on its apparent role as a marker of metastasis. In addition, silibinin inhibits cancer growth in vitro and in PDTX models. Furthermore, metastasis-associated protein 1 (MTA1) could regulate the expression for JMJD5 and had a positive correlation with JMJD5. Moreover, silibinin could downregulate JMJD5 and MTA1 in oral cancer. Present study thus identifies that JMJD5 might be an essential prognostic indicator and therapeutic target against OSCC progression. In addition, silibinin is a potential candidate among novel chemotherapeutic agents or adjuvants for modulating JMJD5 in OSCC, through a mechanism likely involving MTA1/JMJD5 axis.


Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Histone Demethylases/metabolism , Mouth Neoplasms/pathology , Repressor Proteins/metabolism , Silybin/pharmacology , Trans-Activators/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Female , Gene Expression Regulation, Neoplastic , Histone Demethylases/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Prognosis , Repressor Proteins/genetics , Survival Rate , Trans-Activators/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Mediators Inflamm ; 2020: 1237281, 2020.
Article En | MEDLINE | ID: mdl-32587467

PURPOSE: Interleukin-1α (IL-1α) is a potent cytokine that plays a role in inflammatory arthritis and bone loss. Decoy receptor 3 (DCR3) is an immune modulator of monocytes and macrophages. The aim of this study was to investigate the mechanism of DCR3 in IL-1α-induced osteoclastogenesis. METHODS: We treated murine macrophages with DCR3 during receptor activator of nuclear factor kappa Β ligand- (RANKL-) plus IL-1α-induced osteoclastogenesis to monitor osteoclast formation by tartrate-resistant acid phosphatase (TRAP) staining. Osteoclast activity was assessed using a pit formation assay. The mechanisms of inhibition were studied by biochemical analyses, including RT-PCR, immunofluorescent staining, flow cytometry, an apoptosis assay, immunoblotting, and ELISA. RESULTS: DCR3 suppresses IL-1α-induced osteoclastogenesis in both primary murine bone marrow-derived macrophages (BMM) and RAW264.7 cells as it inhibits bone resorption. DCR3 induces RANKL-treated osteoclast precursor cells to express IL-1α, secretory IL-1ra (sIL-1ra), intracellular IL-1ra (icIL-1ra), reactive oxygen species (ROS), and Fas ligand and to activate IL-1α-induced interleukin-1 receptor-associated kinase 4 (IRAK4). The suppression of DCR3 during RANKL- or IL-1α-induced osteoclastogenesis may be due to the abundant secretion of IL-1ra, accumulation of ROS, and expression of Fas ligand in apoptotic osteoclast precursor cells. CONCLUSIONS: We concluded that there is an inhibitory effect of DCR3 on osteoclastogenesis via ROS accumulation and ROS-induced Fas ligand, IL-1α, and IL-1ra expression. Our results suggested that the upregulation of DCR3 in preosteoclasts might be a therapeutic target in inflammatory IL-1α-induced bone resorption.


Fas Ligand Protein/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/metabolism , Osteoclasts/metabolism , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Death/genetics , Cell Death/physiology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Osteoclasts/cytology , RAW 264.7 Cells , Tartrate-Resistant Acid Phosphatase/metabolism
13.
Cells ; 9(4)2020 04 10.
Article En | MEDLINE | ID: mdl-32290250

Interleukin 26 (IL-26) is a new member of the IL-10 family that is highly expressed in rheumatoid arthritis (RA). However, the functions of IL-26 produced by macrophages in RA have not been elucidated. In the present work, we evaluated the effects and the mechanisms of IL-26 on M1 and M2 macrophage differentiation. Human or mouse macrophage cells were treated with lipopolysaccharides (LPS), interferon gamma (IFNγ), or IL-4 alone or concurrently treated with IL-26 to monitor M1 or M2 macrophage subtypes. The expression level of M1 or M2 macrophage genes was evaluated by reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). The molecular mechanisms of downstream signaling activation during differentiation were investigated by immunoblotting assay. Our results found that IL-26 promoted macrophage cells from CD80+ M1 macrophage differentiation, not from the CD206+ M2 phenotype. The messenger RNA of M1-type macrophage markers tumor necrosis factor alpha (TNFα) and inducible nitric oxide synthase (iNOS) was up-regulated in the IL-26-treated group. Also, the M1-related proinflammatory cytokines TNFα and IL-6 were induced after IL-26 stimulation. Interestingly, IL-10, a cytokine marker of M2 macrophage, was also elevated after IL-26 stimulation. Moreover, the M1-like macrophage stimulated by IL-26 underwent cJUN, nuclear factor kappa B (NF-κB), and signal transducer and activator of transcription 1 (STAT1) activation. Our findings suggested the role of IL-26 in synovial macrophages of active rheumatoid arthritis and provided a new insight into IL-26 as a candidate therapeutic target in rheumatoid arthritis.


Interleukins/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Animals , Cell Differentiation/physiology , Cell Polarity/physiology , Humans , Macrophage Activation , Macrophages/cytology , Mice , Phenotype , Proto-Oncogene Proteins c-jun/metabolism , RAW 264.7 Cells
14.
Immunobiology ; 225(3): 151906, 2020 05.
Article En | MEDLINE | ID: mdl-31987604

Dimethyl sulfoxide (DMSO) was discovered during the 19th century by the German chemical industry. DMSO comprises a highly polar group and two non-polar domains, which render it soluble in both aqueous solutions and organic solutions. Furthermore, DMSO can penetrate the cell membrane of both the mammalian cells and the non-mammalian cells and prevent freeze-thaw injuries to the cells. Thus, it is frequently used for the cryopreservation of cells and tissues for laboratory and clinical applications. In contrast to this traditional application, DMSO has recently been shown to possess immunomodulatory effects, such as immune enhancement, and anti-inflammatory effects in the innate immunity. In addition, DMSO also affects the adaptive immunity by regulating the expression of transcription factors in immune cells. This review briefly summarizes and highlights the roles and immunomodulatory effects of DMSO on the immune system and reveals the future clinical therapeutic potential of DMSO treatment in cancer, in autoimmune diseases and in chronic inflammatory diseases.


Dimethyl Sulfoxide/pharmacology , Immunomodulation/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Membrane Permeability , Cryoprotective Agents , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/therapeutic use , Humans , Immune System/diagnostic imaging , Immune System/immunology , Immune System/metabolism , Immunity, Innate/drug effects , Solubility
15.
Article En | MEDLINE | ID: mdl-31636070

Flaviviruses comprise several medically important viruses, including Japanese encephalitis virus, West Nile virus, dengue virus (DENV), yellow fever virus, and Zika virus (ZIKV). A large outbreak of DENV and ZIKV occurred recently, leading to many cases of illness and death. However, despite decades of effort, we have no clinically specific therapeutic drugs against DENV and ZIKV. Previous studies showed that inflammatory responses play a critical role in dengue and Zika virus pathogenesis. Thus, in this study, we examined a series of novel anti-inflammatory compounds and found that treatment with compound 2d could dose dependently reduce viral protein expression and viral progeny production in HEK-293 and Raw264.7 cells infected with four serotypes of DENV and ZIKV. In addition, considering medication safety, compound 2d could not suppress cyclooxygenase-1 (COX-1) enzymatic activities and thus could prevent the side effect of bleeding. Moreover, compound 2d significantly inhibited COX-2 enzymatic activities and prostaglandin E2 levels, associated with viral replication, compared to results with a selective COX-2 inhibitor, celecoxib. Furthermore, administering 5 mg/kg compound 2d to DENV-2-infected AG129 mice prolonged survival and reduced viremia and serum cytokine levels. Overall, compound 2d showed therapeutic safety and efficacy in vitro and in vivo and could be further developed as a potential therapeutic agent for flavivirus infection.


Anti-Inflammatory Agents/pharmacology , Dengue/drug therapy , Zika Virus Infection/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Celecoxib/pharmacology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dengue/enzymology , Dengue/virology , Dengue Virus/classification , Dengue Virus/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Mice , Mice, 129 Strain , RAW 264.7 Cells , Safety , Serogroup , Treatment Outcome , Virus Replication/drug effects , Zika Virus/drug effects , Zika Virus Infection/enzymology , Zika Virus Infection/virology
16.
Toxicol Appl Pharmacol ; 378: 114641, 2019 09 01.
Article En | MEDLINE | ID: mdl-31254568

Encapsulating peritoneal sclerosis (EPS) is a severe complication of peritoneal dialysis (PD). This disease leads to intestinal obstruction with or without peritonitis. The imbalance between the populations of Th17 and regulatory T (Treg) cells (higher Th17 cells and lower Treg cells) is part of the pathogenesis of EPS formation. We demonstrated that dimethyl sulfoxide (DMSO) effectively inhibited autoimmune diabetes recurrence in the islet transplantation of NOD mice via the induction of the differentiation of Treg cells. In this study, we investigated the therapeutic potential of DMSO in the inhibition of EPS formation by a mouse model. Under DMSO treatment, the thickening of the parietal and visceral peritoneum was significantly reduced. The populations of CD4, CD8, and IFN-γ-producing CD4 and CD8 T cells were decreased. The populations of IL-4-producing CD4 T lymphocytes, IL-10-producing CD4 T lymphocytes, CD4 CD69 T lymphocytes and Treg lymphocytes were increased. The expression levels of the cytokines IFN-γ, IL-17a, TNF-α and IL-23, in ascites, were significantly decreased following the DMSO treatment. Furthermore, the differentiation of Treg cells was induced by DMSO from naïve CD4 T cells in vitro, and these cells were adoptively transferred into the EPS mice and significantly prevented EPS formation, exhibiting a comparable effect to the in vivo DMSO treatment. We also demonstrated that the differentiation of Treg cells by DMSO occurred via the activation of STAT5 by its epigenetic effect, without altering the PI3K-AKT-mTOR or Raf-ERK pathways. Our results demonstrated, for the first time, that in vivo DMSO treatment suppresses EPS formation in a mouse model. Furthermore, the adoptive transfer of Treg cells that were differentiated from naïve CD4 T cells by an in vitro DMSO treatment exhibited a similar effect to the in vivo DMSO treatment for the prevention of EPS formation.


Dimethyl Sulfoxide/immunology , Peritoneal Fibrosis/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer/methods , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cytokines/immunology , Diabetes Mellitus, Type 1/immunology , Female , Interleukin-17/immunology , Islets of Langerhans Transplantation/methods , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Phosphatidylinositol 3-Kinases/immunology , Th17 Cells/immunology
17.
Int J Mol Sci ; 20(6)2019 Mar 16.
Article En | MEDLINE | ID: mdl-30884802

Rheumatoid arthritis (RA) is a chronic, inflammatory autoimmune disease of unknown etiology. It is characterized by the presence of rheumatoid factor and anticitrullinated peptide antibodies. The orchestra of the inflammatory process among various immune cells, cytokines, chemokines, proteases, matrix metalloproteinases (MMPs), and reactive oxidative stress play critical immunopathologic roles in the inflammatory cascade of the joint environment, leading to clinical impairment and RA. With the growing understanding of the immunopathogenic mechanisms, increasingly novel marked and potential biologic agents have merged for the treatment of RA in recent years. In this review, we focus on the current understanding of pathogenic mechanisms, highlight novel biologic disease-modifying antirheumatic drugs (DMRADs), targeted synthetic DMRADs, and immune-modulating agents, and identify the applicable immune-mediated therapeutic strategies of the near future. In conclusion, new therapeutic approaches are emerging through a better understanding of the immunopathophysiology of RA, which is improving disease outcomes better than ever.


Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/therapy , Immunomodulation , Inflammation/therapy , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Chemokines/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/immunology , Rheumatoid Factor/immunology
18.
Head Neck ; 41(5): 1260-1269, 2019 05.
Article En | MEDLINE | ID: mdl-30537218

BACKGROUND: Aberrant expression of decoy receptor 3 (DcR3) is considered to be a diagnostic and therapeutic target for human cancers. The aim of this study was to assess DcR3 as a target of the anticancer effects of triptolide (TPL) in preclinical patient-derived tumor xenograft (PDTX) models of oral squamous cell carcinoma (OSCC). METHODS: The expression of DcR3 was evaluated through immunohistochemistry, and correlations were examined using clinical variables. The effects of TPL on the expression of DcR3 and cell proliferation were investigated in OSCC cell lines and in PDTX models. RESULTS: DcR3 overexpression was associated with overall survival and tumor size. TPL significantly decreased tumor growth. Moreover, TPL inhibited the expression of metastasis-associated protein 1 (MTA1), a transcription factor for DcR3 in vivo, in vitro, and in PDTX models. CONCLUSION: TPL appeared to exert anticancer effects by repressing DcR3 and MTA1 in vitro, in vivo, and in PDTX models.


Antineoplastic Agents, Alkylating/pharmacology , Carcinoma, Squamous Cell/metabolism , Diterpenes/pharmacology , Mouth Neoplasms/metabolism , Phenanthrenes/pharmacology , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Diterpenes/therapeutic use , Down-Regulation , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Phenanthrenes/therapeutic use , Xenograft Model Antitumor Assays
19.
BMC Complement Altern Med ; 17(1): 555, 2017 Dec 29.
Article En | MEDLINE | ID: mdl-29284481

BACKGROUND: Danshen is a common traditional Chinese medicine used to treat neoplastic and chronic inflammatory diseases in China. However, the effects of Danshen on human oral cancer cells remain relatively unknown. This study investigated the antiproliferative effects of a Danshen extract on human oral cancer SAS, SCC25, OEC-M1, and KB drug-resistant cell lines and elucidated the possible underlying mechanism. METHODS: We investigated the anticancer potential of the Danshen extract in human oral cancer cell lines and an in vivo oral cancer xenograft mouse model. The expression of apoptosis-related molecules was evaluated through Western blotting, and the concentration of in vivo apoptotic markers was measured using immunohistochemical staining. The antitumor effects of 5-fluorouracil and the Danshen extract were compared. RESULTS: Cell proliferation assays revealed that the Danshen extract strongly inhibited oral cancer cell proliferation. Cell morphology studies revealed that the Danshen extract inhibited the growth of SAS, SCC25, and OEC-M1 cells by inducing apoptosis. The Flow cytometric analysis indicated that the Danshen extract induced cell cycle G0/G1 arrest. Immunoblotting analysis for the expression of active caspase-3 and X-linked inhibitor of apoptosis protein indicated that Danshen extract-induced apoptosis in human oral cancer SAS cells was mediated through the caspase pathway. Moreover, the Danshen extract significantly inhibited growth in the SAS xenograft mouse model. Furthermore, the Danshen extract circumvented drug resistance in KB drug-resistant oral cancer cells. CONCLUSION: The study results suggest that the Danshen extract could be a potential anticancer agent in oral cancer treatment.


Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Drugs, Chinese Herbal/pharmacology , Mouth Neoplasms/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Mice , Mice, SCID , Salvia miltiorrhiza , Xenograft Model Antitumor Assays
20.
Diabetologia ; 60(12): 2409-2417, 2017 12.
Article En | MEDLINE | ID: mdl-28929188

AIMS/HYPOTHESIS: The relative contribution of T helper (Th)1 and Th17 cells in graft rejection is inconclusive, on the basis of evidence provided by different T cell-related cytokine-deficient animal models and graft types. METHODS: We used novel antigen-presenting-cell-specific Il-12p35 (also known as Il12a)-knockout (KO), IL-23p19-knockdown (KD) and IL-27p28-KD strategies to investigate T cell differentiation in islet graft rejection. RESULTS: In vitro dendritic cell-T cell coculture experiments revealed that dendritic cells from Il-12p35-KO and IL-23p19-KD mice showed reduced ability to stimulate IFN-γ and IL-17 production in T cells, respectively. To further explore the T cell responses in islet graft rejection, we transplanted islets into streptozotocin-induced diabetic NOD/severe combined immunodeficiency (SCID) recipient mice with IL-12-, IL-23-, or IL-27-deficient backgrounds and then challenged them with NOD.BDC2.5 T cells. The survival of islet grafts was significantly prolonged in Il-12p35-KO and IL-23p19-KD recipients compared with the control recipients. T cell infiltrations and Th1 cell populations were also decreased in the grafts, correlating with prolonged graft survival. CONCLUSIONS/INTERPRETATION: Our results suggest that IL-12 and IL-23 promote and/or maintain Th1 cell-mediated islet graft rejection. Thus, blockade of IL-12 and IL-23 might act as therapeutic strategies for reducing rejection responses.


Interleukin-12/metabolism , Islets of Langerhans Transplantation/immunology , Animals , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft Rejection/immunology , Graft Survival/immunology , Graft Survival/physiology , Immunohistochemistry , Interleukin-23/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Th17 Cells/enzymology , Th17 Cells/metabolism
...