Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 42
1.
Int Immunopharmacol ; 133: 112145, 2024 May 30.
Article En | MEDLINE | ID: mdl-38691920

Treatment strategies for paediatric neuroblastoma as well as many other cancers are limited by the unfavourable tumour microenvironment (TME). In this study, the TMEs of neuroblastoma were grouped by their genetic signatures into four distinct subtypes: immune enriched, immune desert, non-proliferative and fibrotic. An Immune Score and a Proliferation Score were constructed based on the molecular features of the subtypes to quantify the immune microenvironment or malignancy degree of cancer cells in neuroblastoma, respectively. The Immune Score correlated with a patient's response to immunotherapy; the Proliferation Score was an independent prognostic biomarker for neuroblastoma and proved to be more accurate than the existing clinical predictors. This double scoring system was further validated and the conserved molecular pattern associated with immune landscape and malignancy degree was confirmed. Axitinib and BI-2536 were confirmed as candidate drugs for neuroblastoma by the double scoring system. Both in vivo and in vitro experiments demonstrated that axitinib-induced pyroptosis of neuroblastoma cells activated anti-tumour immunity and inhibited tumour growth; BI-2536 induced cell cycle arrest at the S phase in neuroblastoma cells. The comprehensive double scoring system of neuroblastoma may predict prognosis and screen for therapeutic strategies which could provide personalized treatments.


Axitinib , Immunotherapy , Neuroblastoma , Tumor Microenvironment , Neuroblastoma/immunology , Neuroblastoma/therapy , Neuroblastoma/pathology , Neuroblastoma/drug therapy , Humans , Tumor Microenvironment/immunology , Prognosis , Animals , Immunotherapy/methods , Cell Line, Tumor , Axitinib/therapeutic use , Child , Male , Female , Child, Preschool , Mice , Infant , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects
2.
Immunopharmacol Immunotoxicol ; 44(4): 594-602, 2022 Aug.
Article En | MEDLINE | ID: mdl-35638564

BACKGROUND: In multiple sclerosis (MS), the imbalance between T helper (Th)-17 cells and regulatory T (Treg) cells are critical in autoimmune central nervous system (CNS) inflammation and demyelination. Experimental autoimmune encephalomyelitis (EAE) is an established mouse MS model and simulates MS at diverse levels. OBJECTIVES: This study aims at investigating the impact of anlotinib on the clinical severity of EAE and CD4+ T cell differentiation. MATERIALS AND METHODS: EAE-induced mice were treated with water (control) or 6 mg/kg anlotinib by gavage daily. At the peak of EAE, histopathological examination and flow cytometry analysis of CNS-infiltrating CD4+ T cells were performed. In vitro differentiation of CD4+ T cells under different conditions was detected by flow cytometry and quantitative real-time PCR. Finally, the impacts of anlotinib on the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and the transcription levels of key genes involved in Th17 and Treg differentiation were tested. RESULTS: Anlotinib attenuated the clinical severity of EAE and changed the frequencies of CNS-infiltrating CD4+ T cell subsets. Anlotinib inhibited the differentiation of Th17 cells in vitro, decreased the phosphorylation of STAT3, and reduced the expression of Rorc. Anlotinib promoted the differentiation of Treg cells and upregulated the expression levels of CD39 and CD73. DISCUSSION AND CONCLUSIONS: Anlotinib alleviated the symptoms of EAE via inhibiting the Th17 cell differentiation and promoting Treg cell differentiation. Our study provides new opportunities for the exploitation of anlotinib as a therapeutic agent for the treatment of MS.


Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Cell Differentiation , Disease Models, Animal , Indoles , Mice , Mice, Inbred C57BL , Quinolines , T-Lymphocytes, Regulatory , Th1 Cells , Th17 Cells
3.
Front Immunol ; 13: 845757, 2022.
Article En | MEDLINE | ID: mdl-35401536

Background: Pyroptosis, as an inflammatory form of cell death, is involved in many physiological and pathological processes. Neuroblastoma is the most common extra-cranial solid tumor in children. In this study, the relationship between pyroptosis and tumor microenvironment in neuroblastoma was systematically studied. Methods: We integrated four datasets of neuroblastomas. Through robust clustering of the mRNA expression profiles of 24 pyroptosis-related genes, a total of three pyroptosis patterns were identified. We then constructed a novel scoring method named as pyroscore to quantify the level of pyroptosis in neuroblastoma. Multi-omics data and single-cell RNA sequencing were used to accurately and comprehensively evaluate the effectiveness of pyroscore. Clinical data sets were used to evaluate the use of pyroscore to predict the responsiveness of immune checkpoint treatment. Results: High pyroscore was associated with good prognosis, immune activation, and increased response to checkpoint blockade immunotherapy. Multivariate Cox analysis revealed that the pyroscore was an independent prognostic biomarker and could increase the accuracy of clinical prediction models. Etoposide, a drug picked up by our analysis, could increase the sensitivity of neuroblastoma cells to pyroptosis. External verification using four cohorts of patients who had received immunotherapy showed that high pyroscore was significantly associated with immunotherapy treatment benefit. Conclusions: Taken together, this study revealed that pyroptosis-related gene network could quantify the response of neuroblastoma to immune checkpoint blockade therapy and prognosis, and it may be helpful for clinical practitioners to choose treatment strategies for neuroblastoma patients.


Gene Regulatory Networks , Neuroblastoma , Child , Humans , Immunotherapy , Neuroblastoma/genetics , Neuroblastoma/therapy , Prognosis , Pyroptosis/genetics , Tumor Microenvironment/genetics
4.
Nat Commun ; 12(1): 4300, 2021 07 14.
Article En | MEDLINE | ID: mdl-34262035

Common fragile sites (CFSs) are specific breakage-prone genomic regions and are present frequently in cancer cells. The (E2-independent) E3 ubiquitin-conjugating enzyme FATS (fragile site-associated tumor suppressor) has antitumor activity in cancer cells, but the function of FATS in immune cells is unknown. Here, we report a function of FATS in tumor development via regulation of tumor immunity. Fats-/- mice show reduced subcutaneous B16 melanoma and H7 pancreatic tumor growth compared with WT controls. The reduced tumor growth in Fats-/- mice is macrophage dependent and is associated with a phenotypic shift of macrophages within the tumor from tumor-promoting M2-like to antitumor M1-like macrophages. In addition, FATS deficiency promotes M1 polarization by stimulating and prolonging NF-κB activation by disrupting NF-κB/IκBα negative feedback loops and indirectly enhances both CD4+ T helper type 1 (Th1) and cytotoxic T lymphocyte (CTL) adaptive immune responses to promote tumor regression. Notably, transfer of Fats-/- macrophages protects mice against B16 melanoma. Together, these data suggest that FATS functions as an immune regulator and is a potential target in cancer immunotherapy.


Cell Cycle Proteins/immunology , Macrophages/immunology , Neoplasms/immunology , Tumor Suppressor Proteins/immunology , Ubiquitin-Conjugating Enzymes/immunology , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Immunotherapy , Macrophage Activation , Mice , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Tumor Suppressor Proteins/genetics , Ubiquitin-Conjugating Enzymes/genetics
5.
Front Immunol ; 12: 620238, 2021.
Article En | MEDLINE | ID: mdl-33717117

Pin1 is the only known peptidyl-prolyl cis-trans isomerase (PPIase) that can specifically recognize and isomerize the phosphorylated Serine/Threonine-Proline (pSer/Thr-Pro) motif, change the conformation of proteins through protein phosphorylation, thus regulate various cellular processes in the body. Pin1 plays an important role in cancer, Alzheimer's disease, and autoimmune diseases. However, the specific mechanism of Pin1 regulation in LPS-induced septic shock is unclear. Here, we found that lack of Pin1 reduced shock mortality and organ damage in mice, and NLRP3 inflammasome activation also was reduced in this process. We further confirmed that Pin1 can affect the expression of NLRP3, ASC, Caspase1, and this process can be regulated through the p38 MAPK pathway. We analyzed that p38 MAPK signaling pathway was highly expressed in septic shock and showed a positive correlation with Pin1 in the Gene Expression Omnibus database. We found that Pin1 could affect the phosphorylation of p38 MAPK, have no obvious difference in extracellular signal-regulated kinases (ERK) and Jun-amino-terminal kinase (JNK) signaling. We further found that Pin1 and p-p38 MAPK interacted, but not directly. In addition, Pin1 deficiency inhibited the cleavage of gasdermin D (GSDMD) and promoted the death of macrophages with LPS treatment, and reduced secretion of inflammatory cytokines including IL-1ß and IL-18. In general, our results suggest that Pin1 regulates the NLRP3 inflammasome activation by p38 MAPK signaling pathway in macrophages. Thus, Pin1 may be a potential target for the treatment of inflammatory diseases such as septic shock.


Inflammasomes/metabolism , MAP Kinase Signaling System , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Shock, Septic/etiology , Shock, Septic/metabolism , Animals , Biomarkers , Biopsy , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Inflammation Mediators/metabolism , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Models, Biological , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phosphorylation , Proteolysis , Shock, Septic/pathology
6.
Food Sci Nutr ; 8(10): 5443-5451, 2020 Oct.
Article En | MEDLINE | ID: mdl-33133547

Satsuma (Citrus unshiu Marc.) is rich in high levels of nutrients and popular for its unique flavor, but the consumption of satsuma is limited by some adverse reactions in human body. Previous studies have mainly focused on the effects of storage temperature on the postharvest quality of satsumas, and little attention has paid to the effect of postharvest satsumas on human body immunoregulation. The purpose of this study was to explore the differences in fruit quality, and the effect of satsuma fruits stored at different temperatures on human health. Satsumas stored at low temperature (5.8°C, LT) and room temperature (23 ± 2°C, RT) for 60 days were sampled every 10 days to measure the fruit quality. Sixty volunteers were recruited for the oral stimulation experiment of satsumas, and then the effect of satsumas on human health was examined through the immunoregulation of RAW 264.7 macrophages. The results showed that compared with RT treatment, LT treatment could delay the degradation of satsuma fruit quality. Both the results of the volunteer experiment and cell experiment indicated that postharvest temperature treatments could reduce the adverse effects of satsuma fruits on human body. These findings indicated that 10-day storage at room temperature plus subsequent storage at low temperature was the optimal treatment to maintain fruit quality and functional components of postharvest satsumas. This study provides useful information on satsuma consumption and research work from the perspective of immunoregulation evaluation.

7.
PLoS One ; 15(7): e0235614, 2020.
Article En | MEDLINE | ID: mdl-32678819

Both MicroRNAs and HMGB1 took part in pathological process of myocardial I/R injury though several signaling pathways. We hypothesized that mircoRNA451 (miR-451), a group of small non-coding RNAs, could improve this injury by inhibiting HMGB1. Male SD rats were randomly distributed into 5 groups and subjected to I/R process. After 24 hours of reperfusion injury, the serum content of CK and LDH, the content of MDA in tissue and activity of SOD were detected; The infarcted areas were defined by TTC staining and Evans Blue; TUNEL staining and cleaved-Caspase 3 were used to test apoptosis; HMGB1 was detected by real-time fluorescence quantitative PCR and Western Blotting. Compared with the I/R and I/R+Ad-GFP group, upregulation of miR-451 could reduce the infarcted areas, cardiomyocytes apoptosis index, expression of cleaved-caspase 3 and content of CK and LDH significantly(P<0.05); Meanwhile, upregulation of miR-451 could also obviously inhibit HMGB1, the increase of MDA and the decrease of SOD (P<0.05). So this study revealed that upregulation of miR-451 could prevent myocardial I/R injury by suppressing HMGB1.


HMGB1 Protein/genetics , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Up-Regulation , Animals , Apoptosis/genetics , HMGB1 Protein/metabolism , Male , Malondialdehyde/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
8.
Cardiovasc Toxicol ; 20(6): 618-626, 2020 12.
Article En | MEDLINE | ID: mdl-32632848

Endoplasmic reticulum stress (ERS) is involved in a variety of diseases. Recently, it was found that ERS induces not only apoptosis but also autophagy. Previous studies showed that inhibition of autophagy alleviates cell injury. The purpose of our study was to investigate the involvement of the R-like ER kinase (PERK) in ERS-induced autophagy in H9c2 cardiomyoblasts. To address this aim, therefore, H9c2 cells were treated with PERK agonist and inhibitor after establishment of rapamycin-induced ERS models in H9c2 cardiomyoblasts. Transmission electron microscopy and immunofluorescence staining were used to detect degrees of ERS-induced autophagy, apoptosis and myocardial fibrosis. Western blotting was employed to detect the levels of total and phosphorylated PERK, light chain 3 (LC3), P62, Caspase3, Bcl2 and Bax. Immunofluorescence staining was used to assess α-SMA density. TGF-ß induced H9c2 cardiomyoblasts time-dependently upregulated col I, col III, FN, and LC3 expressions, PERK phosphorylation and α-SMA density, and downregulated P62 level compared with control cells. Treatment with PERK agonist and inhibitor respectively increased and decreased LC3 expression, conversely in P62 level, which is consistent with effect of ERS agonists and inhibitors. And a PERK inhibitor upregulated the expressions of Caspase3 and Bax, and downregulated Bcl2 level, which developed H9c2 cardiomyoblasts. Moreover, siRNA-mediated knockdown of PERK reduced ERS mediated autophagy activity and increased cells apoptosis. On the other hand, elevated autophagy activity could downregulated PERK level. Our finding showed that PERK activity mediates upregulation of ERS-induced autophagy and regulation of cardiomyocyte apoptosis in H9c2 cardiomyoblasts.


Apoptosis/drug effects , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Myocytes, Cardiac/drug effects , Sirolimus/toxicity , eIF-2 Kinase/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Butadienes/pharmacology , Cell Line , Fibrosis , Humans , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Nitriles/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Signal Transduction , eIF-2 Kinase/antagonists & inhibitors
9.
J Immunother Cancer ; 8(1)2020 06.
Article En | MEDLINE | ID: mdl-32503944

BACKGROUND: Immune checkpoint blockade has emerged as a potential cancer immunotherapy. The "don't eat me" signal CD47 in cancer cells binds signal regulatory protein-α on macrophages and prevents their phagocytosis. The role of miR-340 in pancreatic ductal adenocarcinoma (PDAC), especially in tumor immunity, has not been explored. Here, we examined the clinical and biological relevance of miR-340 and the molecular pathways regulated by miR-340 in PDAC. METHODS: CD47 and miR-340 expression and the relationship with cancer patient survival were analyzed by bioinformatics. The mechanism of miR-340 action was explored through bioinformatics, luciferase reporter, qRT-PCR and western blot analyses. The effects of miR-340 on cancer cells were analyzed in terms of apoptosis, proliferation, migration and phagocytosis by macrophages. In vivo tumorigenesis was studied in orthotopic and subcutaneous models, and immune cells from the peripheral and tumor immune microenvironments were analyzed by flow cytometry. Depletion of macrophages was used to verify the role of macrophages in impacting the function of miR-340 in tumor progression. RESULTS: miR-340 directly regulates and inversely correlates with CD47, and it predicts patient survival in PDAC. The restoration of miR-340 expression in pancreatic cancer cells was sufficient to downregulate CD47 and promote phagocytosis of macrophages, further inhibiting tumor growth. The overexpression of miR-340 promoted macrophages to become M1-like phenotype polarized in peripheral and tumor immune microenvironments and increased T cells, especially CD8+ T cells, contributing to the antitumor effect of miR-340. CONCLUSIONS: miR-340 is a key regulator of phagocytosis and antitumor immunity, and it could offer a new opportunity for immunotherapy for PDAC.


CD47 Antigen/metabolism , Carcinoma, Pancreatic Ductal/immunology , Macrophages/immunology , MicroRNAs/genetics , Pancreatic Neoplasms/immunology , Phagocytosis , Tumor Microenvironment/immunology , Animals , Apoptosis , CD47 Antigen/genetics , CD47 Antigen/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Cell Proliferation , Humans , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Prognosis , Survival Rate , Xenograft Model Antitumor Assays
10.
Cell Death Dis ; 10(6): 461, 2019 06 12.
Article En | MEDLINE | ID: mdl-31189875

miR-21 is aberrantly expressed, and plays a role in various types of tumors and many other diseases. However, the mechanism of miR-21 in LPS-induced septic shock is still unclear. In this study, we investigated the mechanism of miR-21 in LPS-induced pyroptosis and septic shock. Here, we show that miR-21 deficiency inhibited NLRP3, ASC, and caspase-1 expression, as well as inflammasome activation in myeloid cells from both mice and humans. We found that the NF-κB pathway was regulated by miR-21, and that A20 was a direct target of miR-21. Furthermore, miR-21 deficiency inhibited the ASC pyroptosome, which restrained caspase-1 activation and GSDMD cleavage, thereby preventing LPS-induced pyroptosis and septic shock. miR-21 deficiency resulted in an increase in A20, which led to decreased IL-1ß production and caspase-1 activation. Caspase-1-mediated GSDMD cleavage was consequently decreased, which prevented pyroptosis in LPS-induced sepsis in mice. Our results demonstrate that miR-21 is a critical positive regulator of the NF-κB pathway and NLRP3 inflammasomes in pyroptosis and septic shock via A20. In addition, by analyzing published miRNA expression profiles in the Gene Expression Omnibus database, we found that the miR-21 levels in peripheral blood from patients with septic shock were elevated. Thus, miR-21 may serve as a potential treatment target in patients with septic shock.


Inflammasomes/metabolism , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/genetics , Shock, Septic/metabolism , Animals , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Cytokines/metabolism , Female , Humans , Inflammasomes/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pyroptosis/drug effects , Shock, Septic/chemically induced , Shock, Septic/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
11.
J Cell Biochem ; 120(9): 15695-15708, 2019 09.
Article En | MEDLINE | ID: mdl-31144365

Muscle redox disturbances and oxidative stress have emerged as a common pathogenetic mechanism and potential therapeutic intervention in some muscle diseases. Parthenolide (PTL), a sesquiterpene lactone found in large amounts in the leaves of feverfew, possesses anti-inflammatory, anti-migraine, and anticancer properties. Although PTL was reported to alleviate cancer cachexia and improve skeletal muscle characteristics in a cancer cachexia model, its actions on oxidative stress-induced damage in C2C12 myoblasts have not been reported and the regulatory mechanisms have not yet been defined. In our study, PTL attenuated H2 O2 -induced growth inhibition and morphological changes. Furthermore, PTL exhibited scavenging activity against reactive oxygen species and protected C2C12 cells from apoptosis in response to H2 O2 . Meanwhile, PTL suppressed collapse of the mitochondrial membrane potential, thereby contributing to normalizing H2 O2 -induced autophagy flux and mitophagy, correlating with inhibiting degradation of mitochondrial marker protein TIM23, the increase in LC3-II expression and the reduction of mitochondria DNA. Besides its protective effect on mitochondria, PTL also prevented H2 O2 -induced lysosomes damage in C2C12 cells. In addition, the phosphorylation of p53, cathepsin B, and Bax/Bcl-2 protein levels, and the translocation of Bax from the cytosol to mitochondria induced by H2 O2 in C2C12 cells was significantly reduced by PTL. In conclusion, PTL modulates oxidative stress-induced mitophagy and protects C2C12 myoblasts against apoptosis, suggesting a potential protective effect against oxidative stress-associated skeletal muscle diseases.


Mitophagy/drug effects , Muscular Diseases/drug therapy , Oxidative Stress/drug effects , Sesquiterpenes/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/drug effects , Humans , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Mitophagy/genetics , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Myoblasts/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
12.
Cell Death Differ ; 26(1): 130-145, 2019 01.
Article En | MEDLINE | ID: mdl-29666475

Inflammasome activation plays key roles in host defense, but also contributes to the pathogenesis of auto-inflammatory, and neurodegenerative diseases. As autophagy is connected with both the innate and adaptive immune systems, autophagic dysfunction is also closely related to inflammation, infection, and neurodegeneration. Here we identify that lincRNA-Cox2, previously known as a mediator of both the activation and repression of immune genes expression in innate immune cells, could bind NF-κB p65 and promote its nuclear translocation and transcription, modulating the expression of inflammasome sensor NLRP3 and adaptor ASC. Knockdown of lincRNA-Cox2 inhibited the inflammasome activation and prevented the lincRNA-Cox2-triggered caspase-1 activation, leading to decreased IL-1ß secretion and weakened TIR-domain-containing adapter-inducing interferon-ß (TRIF) cleavage, thereby enhancing TRIF-mediated autophagy. Elucidation of the link between lincRNA-Cox2 and the inflammasome-autophagy crosstalk in macrophage and microglia reveals a role for lncRNAs in activation of NLRP3 inflammasome and autophagy, and provides new opportunities for therapeutic intervention in neuroinflammation-dependent diseases.


Autophagy/genetics , Inflammasomes/metabolism , Macrophages/metabolism , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA, Long Noncoding/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Autophagy/immunology , Autophagy-Related Protein 5/metabolism , Caspase 1/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , HEK293 Cells , Humans , Inflammasomes/immunology , Inflammation/genetics , Inflammation/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Microglia/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , RNA, Long Noncoding/genetics , Transcription Factor RelA/metabolism
13.
Autophagy ; 15(3): 478-492, 2019 03.
Article En | MEDLINE | ID: mdl-30208760

Microglia are innate immune cells in the central nervous system (CNS), that supplies neurons with key factors for executing autophagosomal/lysosomal functions. Macroautophagy/autophagy is a cellular catabolic process that maintains cell balance in response to stress-related stimulation. Abnormal autophagy occurs with many pathologies, such as cancer, and autoimmune and neurodegenerative diseases. Hence, clarification of the mechanisms of autophagy regulation is of utmost importance. Recently, researchers presented microRNAs (miRNAs) as novel and potent modulators of autophagic activity. Here, we found that Mir223 deficiency significantly ameliorated CNS inflammation, demyelination and the clinical symptoms of experimental autoimmune encephalomyelitis (EAE) and increased resting microglia and autophagy in brain microglial cells. In contrast, the autophagy inhibitor 3-methylademine (3-MA) aggravated the clinical symptoms of EAE in wild-type (WT) and Mir223-deficienct mice. Furthermore, it was confirmed that Mir223 deficiency in mice increased the protein expression of ATG16L1 (autophagy related 16-like 1 [S. cerevisiae]) and LC3-II in bone marrow-derived macrophage cells compared with cells from WT mice. Indeed, the cellular level of Atg16l1 was decreased in BV2 cells upon Mir223 overexpression and increased following the introduction of antagomirs. We also showed that the 3' UTR of Atg16l1 contained functional Mir223-responsive sequences and that overexpression of ATG16L1 returned autophagy to normal levels even in the presence of Mir223 mimics. Collectively, these data indicate that Mir223 is a novel and important regulator of autophagy and that Atg16l1 is a Mir223 target in this process, which may have implications for improving our understanding of the neuroinflammatory process of EAE. Abbreviations: 3-MA: 3-methylademine; ACTB/ß-actin: actin, beta; ATG: autophagy related; ATG16L1: autophagy related 16-like 1 (S. cerevisiae); BECN1: beclin 1, autophagy related; CNR2: cannabinoid receptor 2 (macrophage); CNS: central nervous system; CQ: chloroquine; EAE: experimental autoimmune encephalomyelitis; FOXO3: forkhead box O3; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; H&E: hematoxylin and eosin; ITGAM: integrin alpha M; LPS: lipoplysaccharide; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; miRNAs: microRNAs; MS: multiple sclerosis; PPARG: peroxisome proliferator activated receptor gamma; PTPRC: protein tyrosine phosphatase, receptor type, C; RA: rheumatoid arthritis; SQSTM1: sequestosome 1; TB: tuberculosis; TIMM23: translocase of inner mitochondrial membrane 23; TLR: toll-like receptor.


Autophagy-Related Proteins/metabolism , Autophagy/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , MicroRNAs/metabolism , Microglia/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Proteins/genetics , Cell Line , Central Nervous System/immunology , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Inflammation/metabolism , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Microglia/cytology , Microglia/drug effects , Microscopy, Electron, Transmission , PPAR gamma/genetics , PPAR gamma/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation
14.
Thorac Cancer ; 9(8): 974-988, 2018 08.
Article En | MEDLINE | ID: mdl-29870138

BACKGROUND: This study compared tumor-related signaling pathways with known compounds to determine potential agents for lung adenocarcinoma (LUAD) treatment. METHODS: Kyoto Encyclopedia of Genes and Genomes signaling pathway analyses were performed based on LUAD differentially expressed genes from The Cancer Genome Atlas (TCGA) project and genotype-tissue expression controls. These results were compared to various known compounds using the Connectivity Mapping dataset. The clinical significance of the hub genes identified by overlapping pathway enrichment analysis was further investigated using data mining from multiple sources. A drug-pathway network for LUAD was constructed, and molecular docking was carried out. RESULTS: After the integration of 57 LUAD-related pathways and 35 pathways affected by small molecules, five overlapping pathways were revealed. Among these five pathways, the p53 signaling pathway was the most significant, with CCNB1, CCNB2, CDK1, CDKN2A, and CHEK1 being identified as hub genes. The p53 signaling pathway is implicated as a risk factor for LUAD tumorigenesis and survival. A total of 88 molecules significantly inhibiting the five LUAD-related oncogenic pathways were involved in the LUAD drug-pathway network. Daunorubicin, mycophenolic acid, and pyrvinium could potentially target the hub gene CHEK1 directly. CONCLUSION: Our study highlights the critical pathways that should be targeted in the search for potential LUAD treatments, most importantly, the p53 signaling pathway. Some compounds, such as ciclopirox and AG-028671, may have potential roles for LUAD treatment but require further experimental verification.


Adenocarcinoma of Lung/genetics , Computational Biology/methods , Gene Regulatory Networks/drug effects , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/genetics , Adenocarcinoma of Lung/drug therapy , Computer Simulation , Data Mining , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Docking Simulation , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
15.
Oncol Rep ; 39(5): 2376-2384, 2018 May.
Article En | MEDLINE | ID: mdl-29565453

Bone remodeling can be interrupted by tumor cells which leads to an inappropriate balance of osteoblasts and osteoclasts. As the progenitors of osteoblasts, mesenchymal stem cells (MSCs) have been reported to exhibit an abnormal osteogenic differentiation potential in some cancer­related bone lesions. However, the evidence is very limited in terms of the biological alterations of MSCs in the bone metastasis of non­small cell lung cancers (NSCLC). We investigated the expression and function of miR­139­5p in MSC osteogenic differentiation in vitro in normal and NSCLC-exposed condition. Then, we compared the serum miR­139­5p in stage IV lung adenocarcinoma cancer patients with and without lytic bone metastasis. We found that MSCs exhibited a significant increase in miR­139­5p expression after exposure to osteogenic differentiation induction medium. However, Notch1, which was confirmed as a target of miR­139­5p by luciferase and western blot assays, showed a marked downregulated expression together with its pathway downstream factors during MSC osteogenesis. miR­139­5p positively regulated MSC osteogenic differentiation but this effect was abrogated significantly by Notch1 knockdown. After exposure to conditions of lung cancer cells A549 and L9981, MSCs exhibited significant downregulation of miR­139­5p expression and early osteogenic marker ALP activity. Furthermore, we demonstrated that the expression of serum miR­139­5p from lung adenocarcinoma patients with lytic bone metastasis was significantly lower compared to that in patients with metastases in other organs. The potential roles of miR­139­5p as a biomarker and treatment target in monitoring and controlling bone metastasis in lung cancer patients are worthy of being further explored.


Bone Neoplasms/genetics , Bone Neoplasms/secondary , Down-Regulation , Lung Neoplasms/genetics , MicroRNAs/blood , Receptor, Notch1/genetics , 3' Untranslated Regions , Adult , Aged , Bone Neoplasms/blood , Bone Neoplasms/pathology , Cell Differentiation , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/blood , Lung Neoplasms/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Middle Aged , Neoplasm Staging , Osteolysis , Tumor Cells, Cultured
16.
Biomed Pharmacother ; 102: 464-471, 2018 Jun.
Article En | MEDLINE | ID: mdl-29579707

As a phenylpropanoid and dibenzylbutyrolactone lignan present in medical plants, such as those used in traditional Chinese herbal medicine, including Arctium lappa (Niubang), arctigenin exhibits antimicrobial, anti-inflammatory, and anticancer activities. In this study, we investigated the protective role of arctigenin in Concanavalin A (ConA)-induced acute hepatitis in mice. Arctigenin remarkably reduced the congestion and necroinflammation of livers, and improved hepatic function (ALT and AST) in ConA-induced acute hepatitis in vivo. The infiltration of CD4 T, NKT and macrophages into the livers was found to be reduced with arctigenin treatment. Arctigenin suppressed ConA-induced T lymphocyte proliferations that might have resulted from enhanced IL-10 production by macrophages and CD4 T cells. These results suggested that arctigenin could be a powerful drug candidate for acute hepatitis through immune suppression.


Furans/pharmacology , Hepatitis/complications , Hepatitis/immunology , Lignans/pharmacology , Liver/immunology , Liver/injuries , Protective Agents/pharmacology , Acute Disease , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Concanavalin A , Hepatitis/drug therapy , Inflammation/drug therapy , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-10/biosynthesis , Interleukin-10/genetics , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , RAW 264.7 Cells
17.
Brain Behav Immun ; 70: 305-314, 2018 05.
Article En | MEDLINE | ID: mdl-29545117

Among T helper (Th) cell subsets differentiated from naive CD4+ T cells, IL-17-producing Th17 cells are closely associated with the pathogenesis of autoimmune diseases, including multiple sclerosis (MS) and the MS animal model, experimental autoimmune encephalomyelitis (EAE). The modulation of Th17 differentiation offers a potential avenue for treatment. Although a series of microRNAs (miRNAs) that modulate autoimmune disease development have been reported, further studies on miRNA roles in Th17 differentiation and MS pathogenesis are still warranted. Here, we demonstrated that mice with miR-181c knockdown presented with delayed EAE and slowed disease progression, along with a decreased Th17 cell population. We also found that miR-181c was a Th17 cell-associated miRNA and that Smad7, a negative regulator of TGF-ß signaling, was a potential target of miR-181c. miR-181c knockdown rendered T cells less sensitive to TGF-ß-induced Smad2/3, enhancing the expression of IL-2 which has been reported to inhibit Th17 cell differentiation. Moreover, through the analysis of published miRNA expression profiles from the Gene Expression Omnibus database, increased miR-181c levels were found in peripheral blood from MS patients. Our results identified a novel miRNA that promotes Th17 cell differentiation and autoimmunity, thus miR-181c may serve as a potential treatment target in patients with MS.


Encephalomyelitis, Autoimmune, Experimental/genetics , MicroRNAs/genetics , MicroRNAs/physiology , Animals , Autoimmunity , Cell Differentiation/genetics , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/genetics , Signal Transduction , Smad2 Protein , Smad3 Protein , Th17 Cells/physiology , Transforming Growth Factor beta
18.
Oncotarget ; 8(59): 99693-99706, 2017 Nov 21.
Article En | MEDLINE | ID: mdl-29245934

Pancreatic cancer is an aggressive malignancy that is unresponsive to conventional radiation and chemotherapy. Therefore, development of novel immune therapeutic strategies is urgently needed. L-4F, an Apolipoprotein A-I (ApoA-I) mimetic peptide, is engineered to mimic the anti-inflammatory and anti-oxidative functionalities of ApoA-I. In this work, H7 cells were orthotopically implanted in C57BL/6 mice and treated with L-4F. Then, pancreatic cancer progression and the inflammatory microenvironment were investigated in vivo. The cytotoxicity of L-4F toward H7 cells was assessed in vitro. Furthermore, we investigated the effects of L-4F on macrophage polarization by analyzing the polarization and genes of mouse bone marrow-derived macrophages in vitro. The results show that L-4F substantially reduced the tumorigenicity of H7 cells. L-4F inhibited inflammation by reducing the accumulation of inflammatory cells, such as IL-17A-, IL-4-, GM-CSF-, IL-1ß-, and IL-6-producing cells and Th1 and Th17. Notably, L-4F also decreased the percentage of macrophages in tumor tissues, especially M2 macrophages (CD11b+F4/80+CD206+), which was also confirmed in vitro. Additionally, the expression of the M2 marker genes Arg1, MRC1, and CCL22 and the inflammatory genes IL-6, iNOS, and IL-12 was decreased by L-4F, indicating that L-4F prevents M2 type macrophage polarization. However, L-4F could not directly attenuate H7 cell invasion or proliferation and did not induce apoptosis. In addition, L-4F potently down-regulated STAT3, JNK and ERK signaling pathways but not affects the phosphorylation of p38 in RAW 264.7 cells. These results suggest that L-4F exhibits an effective therapeutic effect on pancreatic cancer progression by inhibiting tumor-associated macrophages and inflammation.

19.
Oncotarget ; 8(43): 74178-74187, 2017 Sep 26.
Article En | MEDLINE | ID: mdl-29088777

Esophageal carcinoma (EC) is a malignancy with high metastatic potential. Chromosomal helicase/ATPase DNA binding protein 1-like (CHD1L) gene is a newly identified oncogene located at Chr1q21, and it is amplified in many solid tumors. However, the status of CHD1L protein expression in EC and its clinical significance is uncertain. This study was designed to investigate the significance of CHD1L expression in human EC and its biological function in EC cells. The expression of CHD1L was examined by immunohistochemistry in 191 surgically resected ECs. The associations between CHD1L expression and clinical pathological parameters and the prognostic value of CHD1L were analyzed. Western blot analysis showed that CHD1L was overexpressed in EC cell lines. In addition, positive CHD1L expression was strongly related to advanced clinical stage (P<0.01), and lymph node metastasis (P<0.01) of EC. The Kaplan-Meier curve indicated that high expression of CHD1L may result in poor prognosis of EC patients (P<0.01), and multivariate analysis showed that CHD1L overexpression was an independent predictor of overall survival. Furthermore, suppression of CHD1L in EC cells increased apoptosis and decreased cell proliferation invasion ability. Our results suggest that CHD1L is a target oncogene with the potential to serve as a novel prognostic biomarker in EC pathogenesis.

20.
Sci Rep ; 7(1): 3209, 2017 06 09.
Article En | MEDLINE | ID: mdl-28600493

Adiponectin (APN), also known as apM1, Acrp30, GBP28 and adipoQ, is a circulating hormone that is predominantly produced by adipose tissue. Many pharmacological studies have demonstrated that this protein possesses potent anti-diabetic, anti-atherogenic and anti-inflammatory properties. Although several studies have demonstrated the antioxidative activity of this protein, the regulatory mechanisms have not yet been defined in skeletal muscles. The aim of the present study was to examine the cytoprotective effects of APN against damage induced by oxidative stress in mouse-derived C2C12 myoblasts. APN attenuated H2O2-induced growth inhibition and exhibited scavenging activity against intracellular reactive oxygen species that were induced by H2O2. Furthermore, treating C2C12 cells with APN significantly induced heme oxygenase-1 (HO-1) and nuclear factor-erythroid 2 related factor 2 (Nrf2). APN also suppressed H2O2-induced mitophagy and partially inhibited the colocalization of mitochondria with autophagosomes/lysosomes, correlating with the expression of Pink1 and Parkin and mtDNA. Moreover, APN protected C2C12 myoblasts against oxidative stress-induced apoptosis. Furthermore, APN significantly reduced the mRNA and protein expression levels of Bax. These data suggest that APN has a moderate regulatory role in oxidative stress-induced mitophagy and suppresses apoptosis. These findings demonstrate the antioxidant potential of APN in oxidative stress-associated skeletal muscle diseases.


Adiponectin/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Protective Agents/pharmacology , Adiponectin/chemistry , Adiponectin/genetics , Animals , Antioxidants/chemistry , Cell Survival/drug effects , Cytoprotection/drug effects , Hydrogen Peroxide/toxicity , Mice , Mitophagy/drug effects , Myoblasts/drug effects , Oxidative Stress/drug effects , Protective Agents/chemistry , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
...