Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Toxicol Appl Pharmacol ; 482: 116787, 2024 01.
Article in English | MEDLINE | ID: mdl-38101582

ABSTRACT

Arsenic is a carcinogen and chronic exposure to arsenic increases the risk of many cancers, including lung cancer. However, the underlying mechanism is not clear. Using A/J mice as a model, our previous animal study has shown that chronic arsenic exposure up-regulates PD-L1 on lung tumor cells which interacts with PD-1 on T cells and inhibits T cell anti-tumor function resulting in increased lung tumorigenesis. In a subsequent in vitro study, we further found that arsenic up-regulated PD-L1 by activating STAT3 at tyrosine 705 in lung epithelial cells, and inhibition of STAT3 mitigated arsenic-induced PD-L1 up-regulation. The present study aims to determine whether STAT3 regulates PD-L1 in the lung of A/J mice and the type of cells from which lung tumor develops upon arsenic exposure. For that purpose, a mouse line with STAT3 conditional knockout in alveolar type 2 (AT2) cells was developed. Our results indicate that arsenic exposure up-regulates PD-L1 in AT2 cells through activating STAT3 in A/J mice. Conditional knockout of STAT3 in AT2 cells inhibited arsenic-induced PD-L1 up-regulation and lung tumor formation. Thus, our findings reveal that STAT3 is the upstream regulator of arsenic-induced PD-L1 up-regulation in AT2 cells and the inhibition of T cell anti-tumor function in the lung, and that AT2 cells are sensitive to arsenic exposure and from which arsenic-enhanced lung tumor formation in A/J mice.


Subject(s)
Arsenic , Lung Neoplasms , Mice , Animals , Arsenic/toxicity , Arsenic/metabolism , B7-H1 Antigen/genetics , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Cell Transformation, Neoplastic , Carcinogenesis , Lung/metabolism , Cell Line, Tumor , STAT3 Transcription Factor/metabolism
2.
Chemosphere ; 346: 140532, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37918541

ABSTRACT

Over the past few decades, there has been a consistent decline in semen quality across the globe, with environmental pollution being identified as the primary cause. Among the various contaminants present in the environment, persistent organic pollutants (POPs) have garnered significant attention due to their high toxicity, slow degradation, bio-accumulation, and long-range migration. PCBs, which include 210 congeners, are a crucial type of POPs that are known to have harmful effects on the environment and human health. Among the various PCB congeners, 3,3',4,4',5-pentachlorobiphenyl (PCB126) is a typical environmental endocrine-disrupting chemical that is widely distributed and has been associated with several health hazards. However, the impact and mechanism of PCB126 on human sperm function has not been fully elucidated. We aimed to investigate the effects of different concentrations of PCB126 (0.01, 0.1, 1, 10 µg/mL) on sperm motility, viability, hyperactivation, and acrosome reaction after incubation for different periods (1 and 2 h), delving deeper into the molecular mechanism of human sperm dysfunction caused by PCB126. First, we investigated the link between PCB126 treatment and the occurrence of protein modifications that are critical to sperm function regulation, such as tyrosine phosphorylation and lysine glutarylation. Second, we examined the potential impact of PCB126 on different parameters related to mitochondrial function, including reactive oxygen species, malondialdehyde levels, mitochondrial membrane potential, mitochondria respiration and adenosine triphosphate generation. Our findings indicate that exposure to environmental pollutants such as PCB126 in vitro may have a negative impact on human sperm functions by interfering with post-translational modifications and mitochondrial functions.


Subject(s)
Environmental Pollutants , Polychlorinated Biphenyls , Humans , Male , Polychlorinated Biphenyls/toxicity , Semen Analysis , Sperm Motility , Semen , Environmental Pollutants/toxicity , Spermatozoa , Protein Processing, Post-Translational , Mitochondria
3.
J Nanobiotechnology ; 21(1): 410, 2023 Nov 06.
Article in English | MEDLINE | ID: mdl-37932752

ABSTRACT

BACKGROUND: Pancreatic cancer is a highly aggressive malignancy with limited treatment options and a poor prognosis. Trophoblast cell surface antigen 2 (TROP2), a cell surface antigen overexpressed in the tumors of more than half of pancreatic cancer patients, has been identified as a potential target for antibody-drug conjugates (ADCs). Almost all reported TROP2-targeted ADCs are of the IgG type and have been poorly studied in pancreatic cancer. Here, we aimed to develop a novel nanobody-drug conjugate (NDC) targeting TROP2 for the treatment of pancreatic cancer. RESULTS: In this study, we developed a novel TROP2-targeted NDC, HuNbTROP2-HSA-MMAE, for the treatment of TROP2-positive pancreatic cancer. HuNbTROP2-HSA-MMAE is characterized by the use of nanobodies against TROP2 and human serum albumin (HSA) and has a drug-antibody ratio of 1. HuNbTROP2-HSA-MMAE exhibited specific binding to TROP2 and was internalized into tumor cells with high endocytosis efficiency within 5 h, followed by intracellular translocation to lysosomes and release of MMAE to induce cell apoptosis in TROP2-positive pancreatic cancer cells through the caspase-3/9 pathway. In a xenograft model of pancreatic cancer, doses of 0.2 mg/kg and 1 mg/kg HuNbTROP2-HSA-MMAE demonstrated significant antitumor effects, and a dose of 5 mg/kg even eradicated the tumor. CONCLUSION: HuNbTROP2-HSA-MMAE has desirable affinity, internalization efficiency and antitumor activity. It holds significant promise as a potential therapeutic option for the treatment of TROP2-positive pancreatic cancer.


Subject(s)
Immunoconjugates , Pancreatic Neoplasms , Humans , Antigens, Surface , Cell Line, Tumor , Immunoconjugates/chemistry , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays , Animals , Pancreatic Neoplasms
4.
Nutrients ; 15(15)2023 Jul 30.
Article in English | MEDLINE | ID: mdl-37571325

ABSTRACT

The developmental origin of health and disease (DOHaD) hypothesis refers to the adverse effects of suboptimal developmental environments during embryonic and early fetal stages on the long-term health of offspring. Intrauterine metabolic perturbations can profoundly impact organogenesis in offspring, particularly affecting cardiac development and giving rise to potential structural and functional abnormalities. In this discussion, we contemplate the existing understanding regarding the impact of maternal metabolic disorders, such as obesity, diabetes, or undernutrition, on the developmental and functional aspects of the offspring's heart. This influence has the potential to contribute to the susceptibility of offspring to cardiovascular health issues. Alteration in the nutritional milieu can influence mitochondrial function in the developing hearts of offspring, while also serving as signaling molecules that directly modulate gene expression. Moreover, metabolic disorders can exert influence on cardiac development-related genes epigenetically through DNA methylation, levels of histone modifications, microRNA expression, and other factors. However, the comprehensive understanding of the mechanistic underpinnings of these phenomena remains incomplete. Further investigations in this domain hold profound clinical significance, as they can contribute to the enhancement of public health and the prevention of cardiovascular diseases.


Subject(s)
Malnutrition , Metabolic Diseases , Prenatal Exposure Delayed Effects , Humans , Female , Obesity/metabolism , Malnutrition/complications , Heart , DNA Methylation , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects/metabolism
5.
Food Chem Toxicol ; 179: 113991, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37595880

ABSTRACT

Decreased sperm motility is a leading cause of male infertility and persistent organic pollutants are known to contribute significantly to the development of this disease. The effects of organochlorine pesticides such as hexachlorocyclohexane (HCH) on human sperm function and their mechanisms of action have received much attention, but are still not fully understood. Herein, we discovered that HCH has a concentration- and time-dependent inhibitory effect on human sperm motility in vitro. Moreover, HCH could reduce the levels of lysine glutarylation (Kglu) and glucose-6-phosphate dehydrogenase activity in sperm. Meanwhile, HCH could increase reactive oxygen species and thereby lead to mitochondrial depolarization and the down-regulation of adenosine triphosphate levels. In particular, we observed that sodium glutarate (Na-glu), the precursor of glutaryl-CoA, could alleviate the inhibitory effect of HCH on sperm Kglu levels, whereas the ROS scavenger N-acetyl-L-cysteine (NAC) had no effect. Intriguingly, both Na-glu and NAC were able to partially inhibit the HCH-induced increase in sperm ROS levels and impaired sperm motility. In conclusion, we propose that HCH inhibits sperm Kglu, leading to the disruption of mitochondrial energy metabolism, which in turn adversely affects sperm motility.


Subject(s)
Hexachlorocyclohexane , Lysine , Humans , Male , Reactive Oxygen Species , Sperm Motility , Semen , Acetylcysteine , Mitochondria
6.
Eur J Med Res ; 27(1): 201, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36242046

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC), the subtype of breast cancer with the highest mortality rate, shows clinical characteristics of high heterogeneity, aggressiveness, easy recurrence, and poor prognosis, which is due to lack of expression of estrogen, progesterone receptor and human epidermal growth factor receptor 2. Currently, neoadjuvant chemotherapy (NAT) is still the major clinical treatment for triple-negative breast cancer. Chemotherapy drugs can be divided into platinum and non-platinum according to the presence of metal platinum ions in the structure. However, which kind is more suitable for treating TNBC remains to be determined. METHODS: The relevant randomized clinical trials (RCTs) that explore the effectiveness of chemotherapy regimens containing platinum-based drugs (PB) or platinum-free drugs (PF) in treating TNBC patients were retrieved through PubMed, EMBASE, Cochrane Library, CNKI, and other literature platforms, above research findings, were included in the meta-analysis. The incidence of overall remission rate (ORR), pathological complete remission rate (pCR), overall survival (OS), disease-free survival (DFS), progression-free survival (PFS), and adverse events (AE) were compared between the two groups. RESULTS: In this study, 12 clinical trials with a total of 4580 patients were included in the analysis. First, the ORR in 4 RCTs was, PB vs PF = 52% vs 48% (RR = 1.05, 95% CI: 0.91-1.21, P = 0.48); the pCR in 5 RCTs was, PB vs PF = 48% vs 41% (RR = 1.38, 95% CI: 0.88-2.16, P = 0.17). CI: 0.88-2.16, P = 0.17; the other 2 RCTs reported significantly higher DFS and OS rates in the PB group compared with the PF group, with the combined risk ratio for DFS in the PB group RR = 0.22 (95% CI:0.06-0.82, P = 0.015); the combined risk ratio for DFS in the PF group RR = 0.15 (95% CI. 0.04-0.61, P = 0.008); OS rate: PB vs PF = 0.046 vs 0.003; secondly, 2 RCTs showed that for patients with BRCA-mutated TNBC, the pCR rate in the PB and PF groups was 18% vs 26%, 95% CI: 2.4-4.2 vs 4.1-5.1; meanwhile, the median subject in the PB group The median PFS was 3.1 months (95% CI: 2.4-4.2) in the PB group and 4.4 months (95% CI: 4.1-5.1) in the PC group; finally, the results of the clinical adverse effects analysis showed that platinum-containing chemotherapy regimens significantly increased the incidence of adverse effects such as thrombocytopenia and diarrhea compared with non-platinum regimens, while the incidence of adverse effects such as vomiting, nausea, and neutropenia was reduced. The incidence of adverse reactions was reduced. CONCLUSION: Compared with non-platinum drugs, platinum drugs significantly improved clinical treatment effective indexes, such as PCR, ORR, PFS, DFS, and OS rate in the treatment of TNBC patients without BRCA mutant may cause more serious hematological adverse reactions. Accordingly, platinum-based chemotherapy should be provided for TNBC patients according to the patient's special details.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/adverse effects , Estrogens , Humans , Platinum/therapeutic use , Randomized Controlled Trials as Topic , Receptors, Progesterone/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
7.
FEBS Open Bio ; 12(11): 2006-2024, 2022 11.
Article in English | MEDLINE | ID: mdl-36054420

ABSTRACT

Hepatic, biliary and pancreatic cancers are a diverse set of malignancies with poor prognoses. It is possible that common molecular mechanisms are involved in the carcinogenesis of these cancers. Here, we identified LINC01537 and seven protein-coding genes by integrative analysis of transcriptomes of mRNAs, microRNAs and long non-coding RNAs from cholangiocarcinoma, hepatocellular carcinoma and pancreatic adenocarcinoma cohorts in TCGA. A predictive model constructed from seven biomarkers was established to successfully predict the survival rate of patients, which was then further verified in external cohorts. Additionally, patients with high-risk scores in our model were prone to epithelial-mesenchymal transition. Finally, activation of the biomarker PDE2A significantly attenuated migration and epithelial-mesenchymal transition in the HepG2 liver cancer cell line.


Subject(s)
Adenocarcinoma , Liver Neoplasms , Pancreatic Neoplasms , Humans , Gene Regulatory Networks/genetics , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Prognosis , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics
8.
Toxicol Appl Pharmacol ; 435: 115845, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34953898

ABSTRACT

Chronic exposure to arsenic promotes lung cancer. Human studies have identified immunosuppression as a risk factor for cancer development. The immune checkpoint pathway of Programmed cell death 1 ligand (PD-L1) and its receptor (programmed cell death receptor 1, PD-1) is the most studied mechanism of immunosuppression. We have previously shown that prolonged arsenic exposure induced cell transformation of BEAS-2B cells, a human lung epithelial cell line. More recently our study further showed that arsenic induced PD-L1 up-regulation, inhibited T cell effector function, and enhanced lung tumor formation in the mice. In the current study, using arsenic-induced BEAS-2B transformation as a model system we investigated the mechanism underlying PD-L1 up-regulation by arsenic. Our data suggests that Lnc-DC, a long non-coding RNA, and signal transducer and activator of transcription 3 (STAT3) mediates PD-L1 up-regulation by arsenic.


Subject(s)
Arsenic/toxicity , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Animals , Cell Line , Female , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Up-Regulation/drug effects
9.
Toxicol Appl Pharmacol ; 426: 115633, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34166680

ABSTRACT

Chronic exposure to environmental arsenic promotes lung cancer. Emerging evidence indicates that compromised host immunity, particularly T cell anti-tumor immunity, may play a critical role in cancer development. However, there is a knowledge gap in terms of the effects of arsenic exposure on T cell anti-tumor immunity and how that may contribute to arsenic lung carcinogenicity. Immunosuppression has been known as a risk factor for many types of cancer, including lung cancer. The development of cancer indicates the success of immunosuppression and escape of cancer cells from host anti-tumor immunity in which T cells are the major component. The anti-tumor immunity is mainly executed by CD8 cytotoxic T cells through their anti-tumor effector function, which can be regulated by immune checkpoint pathways. Some inhibitory receptors on the T cell membrane and their ligands form these pathways, among which programmed death-1 (PD-1), a T cell inhibitory receptor, and its ligand, programmed death-ligand 1 (PD-L1), are best characterized. A/J mice are naturally sensitive to pulmonary carcinogens, prone to develop spontaneous lung tumors later in life and have been frequently used as an animal model for lung tumorigenesis research. Chronic arsenic administration through drinking water has been shown to enhance tumor formation in the lungs of A/J mice. In the current study, using this mouse model we want to determine whether PD-1/PD-L1 plays a role in arsenic-enhanced lung tumorigenesis. The results showed that prolonged arsenic exposure up-regulated PD-1/PD-L1, increased regulatory T cells (Tregs), decreased CD8/Treg ratio, inhibited T cell antitumor function in the lungs and enhanced lung tumor formation, while inhibition of PD-1/PD-L1 restored CD8/Treg ratio and T cell anti-tumor effector function, and mitigated arsenic-enhanced lung tumorigenesis. In addition, inhibition of PD-1/PD-L1 could be a potential preventive strategy to mitigate the tumorigenic action of chronic arsenic exposure.


Subject(s)
Arsenic/toxicity , B7-H1 Antigen/immunology , Carcinogenesis/immunology , Lung Neoplasms/chemically induced , Programmed Cell Death 1 Receptor/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/drug effects , Female , Immunoglobulin G/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Male , Mice , T-Lymphocytes, Regulatory/immunology
10.
J Biol Chem ; 295(41): 14140-14152, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32690611

ABSTRACT

Transcriptional repressor zinc finger and BTB domain containing 1 (ZBTB1) is required for DNA repair. Because DNA repair defects often underlie genome instability and tumorigenesis, we determined to study the role of ZBTB1 in cancer. In this study, we found that ZBTB1 is down-regulated in breast cancer and this down-regulation is associated with poor outcome of breast cancer patients. ZBTB1 suppresses breast cancer cell proliferation and tumor growth. The majority of breast cancers are estrogen receptor (ER) positive and selective estrogen receptor modulators such as tamoxifen have been widely used in the treatment of these patients. Unfortunately, many patients develop resistance to endocrine therapy. Tamoxifen-resistant cancer cells often exhibit higher HER2 expression and an increase of glycolysis. Our data revealed that ZBTB1 plays a critical role in tamoxifen resistance in vitro and in vivo To see if ZBTB1 regulates HER2 expression, we tested the recruitments of ZBTB1 on HER2 regulatory sequences. We observed that over-expressed ZBTB1 occupies the estrogen receptor α (ERα)-binding site of the HER2 intron in tamoxifen-resistant cells, suppressing tamoxifen-induced transcription. In an effort to identify potential microRNAs (miRNAs) regulating ZBTB1, we found that miR-23b-3p directly targets ZBTB1. MiR-23b-3p regulates HER2 expression and tamoxifen resistance via targeting ZBTB1. Finally, we found that miR-23b-3p/ZBTB1 regulates aerobic glycolysis in tamoxifen-resistant cells. Together, our data demonstrate that ZBTB1 is a tumor suppressor in breast cancer cells and that targeting the miR-23b-3p/ZBTB1 may serve as a potential therapeutic approach for the treatment of tamoxifen resistant breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Receptor, ErbB-2/biosynthesis , Repressor Proteins/metabolism , Tamoxifen/pharmacology , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Female , Glycolysis/genetics , Humans , MCF-7 Cells , Mice , Mice, Nude , Receptor, ErbB-2/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics
11.
Mediators Inflamm ; 2020: 3645157, 2020.
Article in English | MEDLINE | ID: mdl-32148437

ABSTRACT

Myasthenia gravis (MG) is a CD4+ T cell-dependent autoimmune disease resulting from aberrant immune response mediated by circulating autoantibodies at the neuromuscular junction. Intravenous immunoglobulin (IVIg) is an expensive and commonly used immunotherapeutic approach to treat patients with MG. The mechanisms of actions involved in IVIg treatment, however, remain to be investigated. In an effort to examine the roles of various subsets of CD4+ T cells in the periphery blood of MG and uncover the mechanisms that contribute to the therapeutical effects of IVIg, we first demonstrated that a subset of CD4+ T cells, CTLA-4-expressing regulatory T (Treg) cells, were underrepresented and functionally defective in MG patients. The dynamic profiling during the IVIg therapy course further revealed an inverse relationship between the frequency of CTLA-4+ Treg and the quantitative MG (QMG) score that represents disease severity. Our mechanistic studies indicated that IVIg expands CTLA-4-Treg cells via modulating antigen-presenting dendritic cells (DCs). To determine the molecular defects of CTLA-4 in abnormities of Treg in MG patients, we demonstrated hypermethylation at -658 and -793 CpGs of CTLA-4 promoter in MG Tregs. Interestingly, IVIg therapy significantly reduced the methylation level at these two sites in MG patients. Overall, our study may suggest a role of CTLA-4 in functionally defected Treg cells in MG and its actions involved in IVIg therapy.


Subject(s)
CTLA-4 Antigen/metabolism , Myasthenia Gravis/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes/metabolism , Adult , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunoglobulins, Intravenous , Lipopolysaccharide Receptors/metabolism , Male , Middle Aged , Myasthenia Gravis/immunology , Real-Time Polymerase Chain Reaction
13.
PLoS One ; 14(5): e0217181, 2019.
Article in English | MEDLINE | ID: mdl-31120927

ABSTRACT

MiR-873/CDK3 has been shown to play a critical role in ERα signaling and tamoxifen resistance. Thus, targeting this pathway may be a potential therapeutic approach for the treatment of ER positive breast cancer especially tamoxifen resistant subtype. Here we report that Norcantharidin (NCTD), currently used clinically as an ani-cancer drug in China, regulates miR-873/CDK3 axis in breast cancer cells. NCTD decreases the transcriptional activity of ERα but not ERß through the modulation of miR-873/CDK3 axis. We also found that NCTD inhibits cell proliferation and tumor growth and miR-873/CDK3 axis mediates cell proliferation suppression of NCTD. More important, we found that NCTD sensitizes resistant cells to tamoxifen. NCTD inhibits tamoxifen induced the transcriptional activity as well ERα downstream gene expressions in tamoxifen resistant breast cancer cells. In addition, we found that NCTD restores tamoxifen induced recruitments of ERα co-repressors N-CoR and SMRT. Knockdown of miR-873 and overexpression of CDK3 diminish the effect of NCTD on tamoxifen resistance. Our data shows that NCTD regulates ERα signaling and tamoxifen resistance by targeting miR-873/CDK3 axis in breast cancer cells. This study may provide an alternative therapy strategy for tamoxifen resistant breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclin-Dependent Kinase 3/metabolism , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/metabolism , MicroRNAs/genetics , Tamoxifen/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cyclin-Dependent Kinase 3/genetics , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Oncotarget ; 8(13): 21398-21409, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423485

ABSTRACT

The alterations in microenvironment upon chronic arsenic exposure may contribute to arsenic-induced lung carcinogenesis. Immune cells, such as macrophages, play an important role in mediating the microenvironment in the lungs. Macrophages carry out their functions after activation. There are two activation status for macrophages: classical (M1) or alternative (M2); the latter is associated with tumorigenesis. Our previous work showed that long-term arsenic exposure induces transformation of lung epithelial cells. However, the crosstalk between epithelial cells and macrophages upon arsenic exposure has not been investigated. In this study, using a co-culture system in which human lung epithelial cells are cultured with macrophages, we determined that long-term arsenic exposure polarizes macrophages towards M2 status through ROS generation. Co-culture with epithelial cells further enhanced the polarization of macrophages as well as transformation of epithelial cells, while blocking macrophage M2 polarization decreased the transformation. In addition, macrophage M2 polarization decreased autophagy activity, which may account for increased cell transformation of epithelial cells with co-culture of macrophages.


Subject(s)
Arsenic/toxicity , Cell Transformation, Neoplastic/chemically induced , Macrophage Activation/immunology , Respiratory Mucosa/pathology , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Coculture Techniques , Flow Cytometry , Humans , Lung Neoplasms/chemically induced , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/immunology
15.
Mol Pharm ; 14(1): 274-283, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27957861

ABSTRACT

Point mutations in the BCR-ABL1 domain and primitive chronic myelogenous leukemia (CML) cells existing in the bone marrow environment insensitive to tyrosine kinase inhibitors (TKIs) have become two major challenges in the CML therapy. In this study, combined TKI ponatinib and JAK2 inhibitor SAR302503 short-term treatment effectively suppressed growth and promoted apoptosis of BaF3/T315I cells in cytokine-containing medium in vitro. SAR302503 prevented cytokine-dependent resistance to ponatinib via inhibition of JAK2/STAT5 phosphorylation. Codelivery of ponatinib and SAR302503 by active bone-targeted polymeric micellar formulation greatly increased the drug accumulation in medullary cavity. The therapeutic efficacy of bone-targeted formulation was demonstrated in BaF3/T315I cells inoculated murine model with no dose-limited toxicity detectable in health mice. Thus, the intravenous injectable bone-homing ponatinib and SAR302503 micellar formulation represents a promising strategy for the treatment of therapy-resistant CML.


Subject(s)
Bone and Bones/drug effects , Drug Resistance, Neoplasm/drug effects , Imidazoles/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Polymers/administration & dosage , Pyridazines/administration & dosage , Pyrrolidines/administration & dosage , Sulfonamides/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone and Bones/metabolism , Cell Line , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Female , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Inbred BALB C , Micelles , Protein Kinase Inhibitors/administration & dosage
16.
Biol Chem ; 398(3): 331-340, 2017 03.
Article in English | MEDLINE | ID: mdl-27648628

ABSTRACT

Pokemon is a transcriptional repressor that belongs to the POZ and Krüppel (POK) protein family. In this study, we investigated the potential interaction between Pokemon and retinoic acid receptor alpha (RARα) and determined the role of Pokemon in regulation of RARα transcriptional activity in the absence of ligand. We found that Pokemon could directly interact with RARα. Moreover, we demonstrated that Pokemon could decrease the transcriptional activity of RARα in the absence of ligand. Furthermore, we showed that Pokemon could repress the transcriptional activity of RARα by increasing the recruitment of nuclear receptor co-repressor (NCoR) and silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) to the retinoic acid response element (RARE) element. Taken together, these data suggest that Pokemon is a novel partner of RARα that acts as a co-repressor to regulate RARα transcriptional activity in the absence of ligand.

17.
Oncoimmunology ; 5(6): e1160182, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27471628

ABSTRACT

Chronic sterile inflammation has been implicated in the pathogenesis of many cancers, including skin cancer. Chronic arsenic exposure is closely associated with the development of skin cancer. However, there is a lack of understanding how arsenic induces chronic inflammation in the skin. Interleukin-1 family cytokines play a central role in regulating immune and inflammatory response. IL-1α, IL-1ß and IL-18 are three pro-inflammatory cytokines in IL-1 family. Their secretion, especially the secretion of IL-1ß and IL-18, is regulated by inflammasomes which are multi-protein complexes containing sensor proteins, adaptor protein and caspase-1. The data from current study show sub-chronic arsenic exposure activates AIM2 inflammasome which in turn activates caspase-1 and enhances the secretion of IL-1ß and IL-18 in HaCaT cells and the skin of BALB/c mice. In addition, arsenic-promoted activation of AIM2 inflammasome and increase of IL-1ß/IL-18 production are inhibited by PKR inhibitor in HaCaT cells or in the skin of PKR mutant mice, indicating a potential role of PKR in arsenic-induced sterile inflammation.

18.
Mol Neurobiol ; 53(2): 1266-1278, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25875864

ABSTRACT

POK erythroid myeloid ontogenic factor (Pokemon), an important proto-oncoprotein, is a transcriptional repressor that regulates the expression of many genes and plays an important role in tumorigenesis. Resveratrol (RSV), a natural polyphenolic compound, has many beneficial biological effects on health. In this study, we investigated the role of Pokemon in RSV-induced biological effects and the effect of RSV on the expression of Pokemon in glioma cells. We found that overexpression of Pokemon decreased RSV-induced cell apoptosis, senescence, and anti-proliferative effects. Moreover, we showed that RSV could efficiently decrease the activity of the Pokemon promoter and the expression of Pokemon. Meanwhile, RSV also inhibited Sp1 DNA binding activity to the Pokemon promoter; whereas, it did not influence the expression and nuclear translocation of Sp1. In addition, we found that RSV could increase the recruitment of HDAC1, but decreased p300 to the Pokemon promoter. Taken together, all these results extended our understanding on the anti-cancer mechanism of RSV in glioma cells.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , DNA-Binding Proteins/genetics , Glioma/drug therapy , Glioma/genetics , Stilbenes/pharmacology , Stilbenes/therapeutic use , Transcription Factors/genetics , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cellular Senescence/drug effects , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , HEK293 Cells , Histone Deacetylase 1/metabolism , Humans , Promoter Regions, Genetic , Protein Binding/drug effects , Protein Transport/drug effects , Resveratrol , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism
20.
Cerebellum ; 14(4): 386-97, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25592072

ABSTRACT

Ethanol-induced neuronal loss is closely related to the pathogenesis of fetal alcohol spectrum disorders. The cerebellum is one of the brain areas that are most sensitive to ethanol. The mechanism underlying ethanol neurotoxicity remains unclear. Our previous in vitro studies have shown that the double-stranded RNA (dsRNA)-activated protein kinase (PKR) regulates neuronal apoptosis upon ethanol exposure and ethanol activates PKR through association with its intracellular activator RAX. However, the role of PKR and its interaction with RAX in vivo have not been investigated. In the current study, by utilizing N-PKR-/- mice, C57BL/6J mice with a deficient RAX-binding domain in PKR, we determined the critical role of RAX/PKR association in PKR-regulated ethanol neurotoxicity in the developing cerebellum. Our data indicate that while N-PKR-/- mice have a similar BAC profile as wild-type mice, ethanol induces less brain/body mass reduction as well as cerebellar neuronal loss. In addition, ethanol promotes interleukin-1ß (IL-1ß) secretion, and IL-1ß is a master cytokine regulating inflammatory response. Importantly, ethanol-promoted IL-1ß secretion is inhibited in the developing cerebellum of N-PKR-/- mice. Thus, RAX/PKR interaction and PKR activation regulate ethanol neurotoxicity in the developing cerebellum, which may involve ethanol-induced neuroinflammation. Further, PKR could be a possible target for pharmacological intervention to prevent or treat fetal alcohol spectrum disorder (FASD).


Subject(s)
Central Nervous System Depressants/toxicity , Cerebellum , Ethanol/toxicity , Neurotoxicity Syndromes/etiology , eIF-2 Kinase/deficiency , Age Factors , Animals , Animals, Genetically Modified , Animals, Newborn , Apoptosis/drug effects , Apoptosis/genetics , Cells, Cultured , Cerebellum/drug effects , Cerebellum/growth & development , Cerebellum/pathology , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/pathology , Organ Size/drug effects , Organ Size/genetics , Sincalide/pharmacology , Time Factors , eIF-2 Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...