Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Thorax ; 78(4): 402-408, 2023 04.
Article En | MEDLINE | ID: mdl-35301243

BACKGROUND: Birt-Hogg-Dubé syndrome (BHD) is a clinical syndrome manifesting with cystic lung disease and pneumothorax. Features of BHD result from the loss-of-function mutations of the folliculin (FLCN) gene. Chronic obstructive pulmonary disease (COPD), characterised by an irreversible airflow limitation, is primarily caused by cigarette smoking. OBJECTIVE: Given that COPD often shares structural features with BHD, we investigated the link between COPD, cigarette smoke (CS) exposure and FLCN expression. METHODS: We measured the expression of FLCN in human COPD lungs and CS-exposed mouse lungs, as well as in CS extract (CSE)-exposed immortalised human airway epithelial cells by immunoblotting. RESULTS: We found that the lung FLCN protein levels in smokers with COPD and CS exposure mice exhibit a marked decrease compared with smokers without COPD and room air exposure mice, respectively. We confirmed CS induced degradation of FLCN in immortalised human bronchial epithelial Beas-2B cells via ubiquitin proteasome system. Further, siRNA targeting FLCN enhanced CSE-induced cytotoxicity. By contrast, FLCN overexpression protected cells from CSE-induced cytotoxicity. We found that FBXO23, the ubiquitin E3 ligase subunit, specifically binds to and targets FLCN for degradation. Inhibition of ATM (ataxia-telangiectasia mutated) attenuated CSE induced FLCN degradation, suggesting a role of ATM in FLCN proteolysis. We further confirmed that the mutant of major FLCN phosphorylation site serine 62A is resistant to CSE-induced degradation and cytotoxicity. CONCLUSIONS: Our study demonstrates that CS exposure is a secondary cause of FLCN deficiency due to the enhanced proteolysis, which promoted airway epithelial cell death.


Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Animals , Humans , Mice , Cigarette Smoking/adverse effects , Lung/chemistry , Lung/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitins/metabolism
2.
Thorax ; 78(4): 383-393, 2023 04.
Article En | MEDLINE | ID: mdl-35354645

BACKGROUND: One hallmark of sepsis is the reduced number of lymphocytes, termed lymphopenia, that occurs from decreased lymphocyte proliferation or increased cell death contributing to immune suppression. Histone modification enzymes regulate immunity by their epigenetic and non-epigenetic functions; however, the role of these enzymes in lymphopenia remains elusive. METHODS: We used molecular biological approaches to investigate the high expression and function of a chromatin modulator protein arginine N-methyltransferase 4 (PRMT4)/coactivator-associated arginine methyltransferase 1 in human samples from septic patients and cellular and animal septic models. RESULTS: We identified that PRMT4 is elevated systemically in septic patients and experimental sepsis. Gram-negative bacteria and their derived endotoxin lipopolysaccharide (LPS) increased PRMT4 in B and T lymphocytes and THP-1 monocytes. Single-cell RNA sequencing results indicate an increase of PRMT4 gene expression in activated T lymphocytes. Augmented PRMT4 is crucial for inducing lymphocyte apoptosis but not monocyte THP-1 cells. Ectopic expression of PRMT4 protein caused substantial lymphocyte death via caspase 3-mediated cell death signalling, and knockout of PRMT4 abolished LPS-mediated lymphocyte death. PRMT4 inhibition with a small molecule compound attenuated lymphocyte death in complementary models of sepsis. CONCLUSIONS: These findings demonstrate a previously uncharacterised role of a key chromatin modulator in lymphocyte survival that may shed light on devising therapeutic modalities to lessen the severity of septic immunosuppression.


Lymphopenia , Protein-Arginine N-Methyltransferases , Sepsis , Animals , Humans , Arginine/genetics , Caspase 3/genetics , Caspase 3/immunology , Chromatin , Lipopolysaccharides/pharmacology , Lymphopenia/etiology , Lymphopenia/genetics , Lymphopenia/immunology , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Sepsis/complications , Sepsis/genetics , Sepsis/immunology
3.
Cell Death Dis ; 12(9): 828, 2021 09 03.
Article En | MEDLINE | ID: mdl-34480022

Lung epithelial cell death is a prominent feature of acute lung injury and acute respiratory distress syndrome (ALI/ARDS), which results from severe pulmonary infection leading to respiratory failure. Multiple mechanisms are believed to contribute to the death of epithelia; however, limited data propose a role for epigenetic modifiers. In this study, we report that a chromatin modulator protein arginine N-methyltransferase 4/coactivator-associated arginine methyltransferase 1 (PRMT4/CARM1) is elevated in human lung tissues with pneumonia and in experimental lung injury models. Here PRMT4 is normally targeted for its degradation by an E3 ubiquitin ligase, SCFFBXO9, that interacts with PRMT4 via a phosphodegron to ubiquitinate the chromatin modulator at K228 leading to its proteasomal degradation. Bacterial-derived endotoxin reduced levels of SCFFBXO9 thus increasing PRMT4 cellular concentrations linked to epithelial cell death. Elevated PRMT4 protein caused substantial epithelial cell death via caspase 3-mediated cell death signaling, and depletion of PRMT4 abolished LPS-mediated epithelial cell death both in cellular and murine injury models. These findings implicate a unique molecular interaction between SCFFBXO9 and PRMT4 and its regulation by endotoxin that impacts the life span of lung epithelia, which may play a key role in the pathobiology of tissue injury observed during critical respiratory illness.


Endotoxins/toxicity , Epithelial Cells/enzymology , Epithelial Cells/pathology , Lung/pathology , Protein-Arginine N-Methyltransferases/metabolism , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Caspase 3/metabolism , Cell Death/drug effects , Cell Line , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Epithelial Cells/drug effects , F-Box Proteins/metabolism , Humans , Lysine/metabolism , Mice , Models, Biological , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Ubiquitin/metabolism , Ubiquitination/drug effects
4.
Genes (Basel) ; 11(6)2020 06 26.
Article En | MEDLINE | ID: mdl-32604881

In quiescent cells, primary cilia function as a mechanosensor that converts mechanic signals into chemical activities. This unique organelle plays a critical role in restricting mechanistic target of rapamycin complex 1 (mTORC1) signaling, which is essential for quiescent cells to maintain their quiescence. Multiple mechanisms have been identified that mediate the inhibitory effect of primary cilia on mTORC1 signaling. These mechanisms depend on several tumor suppressor proteins localized within the ciliary compartment, including liver kinase B1 (LKB1), AMP-activated protein kinase (AMPK), polycystin-1, and polycystin-2. Conversely, changes in mTORC1 activity are able to affect ciliogenesis and stability indirectly through autophagy. In this review, we summarize recent advances in our understanding of the reciprocal regulation of mTORC1 and primary cilia.


Cilia/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanotransduction, Cellular/genetics , AMP-Activated Protein Kinase Kinases , Autophagy/genetics , Cilia/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Kinases , Protein Serine-Threonine Kinases , Signal Transduction/genetics , TRPP Cation Channels
5.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L252-L263, 2020 02 01.
Article En | MEDLINE | ID: mdl-31746627

Cigarette smoking increases susceptibility for microbial infection in respiratory system. However, the underlying molecular mechanism(s) is not fully elucidated. Here we report that cigarette smoking extract (CSE) increases bacterial load in lung epithelial cells via downregulation of the ubiquitin-specific protease 25 (USP25)/histone deacetylase 11 (HDAC11) axis. CSE treatment decreases HDAC11 at protein level in lung epithelial cells without significant changes of its transcription. Concomitantly, CSE treatment accelerates a ubiquitin-specific protease USP25 ubiquitination and degradation. Coimmunoprecipitation studies showed that USP25 associated with HDAC11. USP25 catalyzes deubiquitination of HDAC11, which regulates HDAC11 protein stability. CSE-mediated degradation of USP25 thereafter reduces HDAC11 at the protein level. Interestingly, CSE-downregulated USP25/HDAC11 axis increases the bacterial load of Pseudomonas aeruginosa in lung epithelial cells. These findings suggest that CSE-downregulated USP25 and HDAC11 may contribute to high susceptibility of bacterial infection in the cigarette smoking population.


Bacterial Load/physiology , Cigarette Smoking/adverse effects , Histone Deacetylases/metabolism , Lung/metabolism , Lung/microbiology , Pseudomonas aeruginosa/physiology , Signal Transduction , Ubiquitin Thiolesterase/metabolism , Animals , Cell Line , Enzyme Stability , Female , Humans , Lysine/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitination
6.
Biochim Biophys Acta Gene Regul Mech ; 1861(9): 773-782, 2018 09.
Article En | MEDLINE | ID: mdl-30745998

The histone acetyltransferase HBO1 (Histone acetyltransferase binding to origin recognition complex 1, Myst2/Kat7) participates in a range of life processes including DNA replication and tumorigenesis. Recent studies revealed that HBO1 is involved in gene transcriptional activation. However, the molecular behavior of HBO1 in inflammation is yet to be studied. Here we report that endotoxin lipopolysaccharide (LPS) elevates HBO1 protein level via up-regulating UPS25 (ubiquitin specific peptidase 25) and alters inflammatory gene transcription in THP-1 monocytes and in human primary macrophages. LPS protects HBO1 from ubiquitin proteasomal degradation without significantly altering its transcription. By immunoprecipitation, we identified that HBO1 associates with a deubiquitinating enzyme USP25 in THP-1 cells. LPS increases protein level of USP25 resulting in accumulation of HBO1 by suppression of HBO1 ubiquitination. Stabilized-HBO1 modulates inflammatory gene transcription in THP-1 cells. These findings indicate that USP25 promotes stability of HBO1 in bacterial infection thereby enhances HBO1-mediated inflammatory gene transcription.


Enzyme Stability/drug effects , Histone Acetyltransferases/metabolism , Lipopolysaccharides/pharmacology , Ubiquitin Thiolesterase/metabolism , Histone Acetyltransferases/genetics , Humans , Macrophages/drug effects , Macrophages/metabolism , THP-1 Cells , Transcription, Genetic/drug effects , Ubiquitin Thiolesterase/genetics , Ubiquitination/drug effects
7.
J Cell Sci ; 130(20): 3578-3587, 2017 Oct 15.
Article En | MEDLINE | ID: mdl-28883095

E3 ubiquitin ligase recognizes its protein substrates via specific molecular signatures for ubiquitin proteasomal degradation. However, the role of acetylation/deacetylation in the process of E3 ubiquitin ligase recognizing its protein substrates is not fully studied. Here, we report that a tandem IK motif in protein arginine methyltransferase 1 (PRMT1) forms an acetyldegron to recruit the F-box/LRR-repeat protein 17 (FBXL17), a component of the SKP1-CUL1-F-box protein (SCF)-type E3 ubiquitin ligase complex. PRMT1 is polyubiquitylated for proteasome degradation with a half-life of approximately 4 h in lung epithelial cells. SCFFbxl17 mediates PRMT1 polyubiquitylation at K117. SCFFbxl17 specifically binds PRMT1 via a unique motif IKxxxIK. Strikingly, the acetylation/deacetylation status of the lysine residues within the motif determines Fbxl17 binding. Deacetylation on both K200 and K205 by Sirtuin 1 (Sirt1) and acetylation of p300 (EP300) on K205 collaboratively prepare the motif for SCFFbxl17 binding thereby triggering PRMT1 protein degradation. Pathogen-derived lipopolysaccharide (LPS) downregulates Sirt1 and p300 to protect PRMT1 from degradation. This study demonstrates that LPS promotes PRMT1 stability by blockade of PRMT1 and SCFFbxl17 binding via an acetylation/deacetylation-modified acetyldegron; and LPS-elevated levels of PRMT1 lead to bronchial epithelial cell overgrowth in pulmonary inflammatory diseases.


F-Box Proteins/metabolism , Lipopolysaccharides/pharmacology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Sirtuin 1/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Motifs , Animals , Binding Sites , Cell Line , Humans , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Proteolysis , Ubiquitination/immunology
8.
Am J Physiol Cell Physiol ; 313(3): C285-C294, 2017 Sep 01.
Article En | MEDLINE | ID: mdl-28637674

Oxidative stress impacts normal cellular function leading to the pathogenesis of various diseases including pulmonary illnesses. Protein arginine methyltransferase 4 (PRMT4) is critical for normal lung alveolar epithelial cell development; however, the regulation of PRMT4 within such pulmonary diseases has yet to be elucidated. Using biochemical approaches, we uncovered that peroxide (H2O2) treatment decreases PRMT4 protein stability in murine lung epithelial (MLE12) cells to impede cell migration. Protein kinase glycogen synthase kinase 3ß (GSK-3ß) interacts with PRMT4 and catalyzes PRMT4 T132 phosphorylation that protects PRMT4 from ubiquitin proteasomal degradation. H2O2 downregulates GSK-3ß to reduce PRMT4 at protein level. PRMT4 promotes cell migration and H2O2 degrades PRMT4 to inhibit lung epithelial cell migration. These observations demonstrate that oxidative stress destabilizes PRMT4 via GSK-3ß signaling to impede lung epithelial cell migration that may hinder the lung repair and regeneration process.


Cell Movement/physiology , Glycogen Synthase Kinase 3 beta/metabolism , Oxidative Stress/physiology , Protein-Arginine N-Methyltransferases/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/physiology , Wound Healing/physiology , Animals , Cell Line , Enzyme Stability , Epithelial Cells/cytology , Epithelial Cells/physiology , Gene Expression Regulation, Enzymologic/physiology , Mice
9.
Mol Carcinog ; 54 Suppl 1: E192-204, 2015 Jun.
Article En | MEDLINE | ID: mdl-25820656

Lung cancer is a major health problem, and is considered one of the deadliest cancers in humans. It is refractory to current treatments, and the mechanisms of lung cancer are unknown. Long noncoding RNAs (lncRNAs) are involved in various biological processes and human diseases. However, the exact functional roles and mechanisms of lncRNAs are largely unclear. In this study, we attempted to identify lung-cancer-related lncRNAs. We found changes in lncRNA expression in the anti-benzo(a) pyrene-7,8-diol-9,10-epoxide (anti-BPDE)-transformed human bronchial epithelial cell line (16HBE-T cells) using microarrays and qRT-PCR. Of these lncRNAs, LOC728228 was upregulated relative to its expression in control untransformed16HBE (16HBE-N) cells. LOC728228 knockdown inhibited cell proliferation, caused G0/G1-phase cell-cycle arrest, reduced cellular migration, suppressed colony formation in vitro, and inhibited tumor growth in a nude mouse xenograft model. LOC728228 knockdown also suppressed cyclin D1 expression, and the depletion of cyclin D1 induced G0/G1-phase cell-cycle arrest and inhibited cell proliferation, thus influencing the malignant potential of cancer cells. In summary, our results suggest that lncRNA LOC728228 has an oncogene-like function and plays a vital role in human lung cancer.


7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Lung Neoplasms/pathology , RNA, Long Noncoding/physiology , Animals , Cell Line, Transformed , Humans , Mice , Mice, Nude , Polymerase Chain Reaction
10.
Toxicology ; 331: 57-65, 2015 May 04.
Article En | MEDLINE | ID: mdl-25772433

Phosphorylation of histone H2AX (γ-H2AX) in response to DNA double-strand breaks (DSBs) should be eliminated from the sites of DNA damage to fulfill the DNA repair and release cells from the growth arrest. Previous study showed that protein phosphatase 2A (PP2A) interact with γ-H2AX that lead to the dephosphorylation of γ-H2AX. Here, we examined the effects of suppression of PP2A regulatory subunits on dephosphorylation of γ-H2AX in human embryonic kidney epithelial cells (HEK) treated by topoisomerase I inhibitor camptothecin (CPT). We found that cells with suppression of B55α or B56ϵ were more sensitive to DNA damage agents. Suppression of B56ϵ led to persistence of γ-H2AX, resulting in prolonged DSBs repair and increased chromatin instability measured by comet assay. In addition, the deficiency of B56ϵ impaired the cell cycle regulation and the DNA repair pathway of homologous recombination (HR). Notably, we detected that PP2A B56ϵ subunit was involved directly in dephosphorylation of γ-H2AX and translocated from cytoplasm to nucleus upon the treatment of CPT. Our findings demonstrate that PP2A holoenzyme containing B56ϵ is responsible for the dephosphorylation of γ-H2AX and regulation of DNA repair of DSBs induced by CPT.


Camptothecin/pharmacology , DNA Breaks, Double-Stranded , DNA Repair , Histones/metabolism , Protein Phosphatase 2/metabolism , Topoisomerase I Inhibitors/pharmacology , Active Transport, Cell Nucleus , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Genomic Instability/drug effects , HEK293 Cells , Humans , Phosphorylation , Protein Phosphatase 2/genetics , RNA Interference , Signal Transduction , Time Factors , Transfection
11.
Int J Clin Exp Pathol ; 7(3): 978-86, 2014.
Article En | MEDLINE | ID: mdl-24696715

Granzyme B and perforin, two of the most important components, have shown anticancer properties in various cancers, but their effects in laryngeal cancer remain unexplored. Here we decided to examine the effects of Granzyme B and perforin in Hep-2 cells and clarify the role of perforin and granzyme B in the tumorigenicity of laryngeal cancer cell line. Hep-2 cells were transfected with pVAX1-PIG co-expression vector (comprising perforin and granzyme B genes), and then the growth and apoptosis of these Hep-2 cells were evaluated. The tumorigenicity of Hep-2 cell line co-expressing perforin and granzyme B genes was tested in BALB/c nu/nu mice. We found that the co-expression of perforin and granzyme B genes could obviously inhibit cell focus formation and induce cell apoptosis in Hep-2 cells. Furthermore, after subcutaneous injection of Hep-2 cells transfected with pVAX1-PIG, an extensive delay in tumor growth was observed in BALB/c-nu/nu mice. Moreover, our studies demonstrated that the anticancer activity of perforin and granzyme B was sustainable in vivo as tumor development by inducing cell apoptosis. Taken together, our data indicate that the co-expression of perforin and granzyme B genes exhibits anticancer potential, and hopefully provide potential therapeutic applications in laryngeal cancer.


Apoptosis/genetics , Carcinogenesis/metabolism , Granzymes/biosynthesis , Laryngeal Neoplasms/metabolism , Perforin/biosynthesis , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Flow Cytometry , Granzymes/genetics , Heterografts , Humans , Immunohistochemistry , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Perforin/genetics , Transfection
12.
Protein Expr Purif ; 95: 38-43, 2014 Mar.
Article En | MEDLINE | ID: mdl-24291445

Granzyme B and perforin, two major effector molecules in the granule-mediated cytolytic pathway, are thought to be involved in suppression of tumor progression. In this study, the pGEX-4T-1 expression vector was used to express full-length human perforin or granzyme B as a GST-tagged fusion protein in Escherichia coli (E. coli). GST-tagged proteins were induced with IPTG and purified by GSTrap 4B columns. Purified fusion proteins migrated at the predicted molecular mass on SDS-PAGE and were recognized by specific antibodies. Moreover, the fusion proteins can induce apoptosis and directly inhibit the growth of human laryngeal cancer Hep-2 cells in vitro. These results suggest that active perforin and granzyme B fusion proteins can be produced in E. coli and exhibit anticancer potential in laryngeal cancer cells.


Antineoplastic Agents/pharmacology , Granzymes/metabolism , Perforin/metabolism , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Escherichia coli/genetics , Glutathione Transferase , Granzymes/genetics , Granzymes/isolation & purification , Humans , Laryngeal Neoplasms , Perforin/genetics , Perforin/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Toxicol Lett ; 223(2): 205-10, 2013 Nov 25.
Article En | MEDLINE | ID: mdl-24084393

It has recently been found that the new class of transcripts, long non-coding RNAs (lncRNAs), are pervasively transcribed in the genome. LncRNAs are a large family of non-coding RNAs and regulate many protein-coding genes. Growing evidence indicates that lncRNAs may play an important functional role in cancer biology. Emerging data have shown that lncRNAs are closely related to the occurrence and development of lung cancer. However, the function and mechanism of lncRNAs in lung cancer remain elusive. Here, we investigated the role of a novel lncRNA in transformed human bronchial epithelial cells induced by benzo(a)pyrene. After establishing the transformed cell model using the BEAS-2B cell line in vitro, we found that expression of lncRNA-DQ786227 was high and changed during the transformation of BEAS-2B cells. Silencing of lncRNA-DQ786227 expression in malignant transformed BEAS-2B cells led to inhibition of cell proliferation and colony formation, and increased apoptosis. LncRNA-DQ786227 dramatically promoted the ability of BEAS-2B-T cells to form colonies in vitro and develop tumors in nude mice. These findings revealed that lncRNA-DQ786227 acts as an oncogene in malignantly transformed BEAS-2B cells induced by benzo(a)pyrene. The identification of lncRNA could provide new insight into the molecular mechanisms of chemical carcinogenesis.


Benzo(a)pyrene/adverse effects , Cell Transformation, Neoplastic/pathology , RNA, Long Noncoding/genetics , Animals , Apoptosis/drug effects , Bronchi/cytology , Bronchi/drug effects , Bronchi/pathology , Carcinogenesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Epithelial Cells , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Mice, Nude , RNA, Long Noncoding/metabolism
14.
Wei Sheng Yan Jiu ; 38(6): 645-8, 2009 Nov.
Article Zh | MEDLINE | ID: mdl-20047213

OBJECTIVE: To study the application of different metabolic activation systems in benzo(a)pyrene [B(a)P]-induced human bronchial epithelium cell HBETR transformation. METHODS: In vitro metabolic activations of B(a)P were compared with rat liver S9 fraction mix, overexpression of a key enzyme (P450 CYP1A1), and prior low dose B(a)P (1 micromol/L) induction. Using soft agar assay and tumorigenicity assay, the different metabolic activation systems were compared to the influence on transformation of human bronchial epithelium cell HBETR. RESULTS: Both immunoblotting and enzyme activity showed that cells overexpressing CYP1A1 (HBETR-1A1) and 48 h after low dose B(a)P induction (HBETR-IN) had high-level expression of CYP1A1. There were no obvious changes in the biology characteristic of these cells. The latencies of cell transformation in HBETR-1A1 and HBETR-IN cells were 11 weeks when cells were treated with B(a)P at concentration of 20 micromol/L, while it took 14 weeks to achieve cell transformation in their control cells. The latencies of malignant transformation in HBETR cells in presence or absence of S9-mix were 14 weeks and 20 weeks, respectively. The efficiencies of cell transformation were in consonance with the protein level of endogenous CYP1A1 enzyme and its enzyme activity. CONCLUSION: The three metabolic conditions of the addition of rat liver S9 fraction mix, overexpression of a key enzyme (CYP1A1), and low dose B(a) P induction could enhance the B(a)P metabolic activation and shorten the latency of malignant transformation. In terms of the feasibility, difficulty of manipulation, stability, and reliability, low dose B(a)P induction could seem to be a prospective system used in metabolic activation in comparison with rat liver S9 fraction mix addition.


Benzo(a)pyrene/toxicity , Biotransformation/drug effects , Bronchi/cytology , Cell Transformation, Neoplastic , Epithelial Cells/cytology , Benzo(a)pyrene/pharmacokinetics , Biotransformation/genetics , Carcinogenicity Tests/methods , Cell Line , Cytochrome P-450 CYP1A1/metabolism , Humans
15.
Zhonghua Yu Fang Yi Xue Za Zhi ; 42(6): 395-9, 2008 Jun.
Article Zh | MEDLINE | ID: mdl-19035039

OBJECTIVE: To establish human bronchial epithelial cell lines over expressing oncogene and to investigate its application in detection of carcinogen-induced cell transformation. METHODS: Mediated by retrovirus infection, human telomerase catalytic subunit, hTERT was introduced into immortal human bronchial epithelial cells (16HBE) and followed by introduction of the oncogenic allele H-Ras(V12), or c-Myc or empty vector, creating cell lines 16HBETR, 16HBETM and 16HBETV, respectively. Biological characteristics of these cell lines including morphology, proliferation, and chromosomal aberration were examined to access whether they were transformed. Soft agar experiment and nude mice subcutaneous injection were performed using pre-transformed 16HBE cells induced by known carcinogens, nickel sulfate (NiSO4) and 7, 8, -dihydrodiol-9, 10-epoxide benzo[a] pyrene (BPDE). RESULTS: With detection of telomerase activity and Western blotting, the expression of target proteins was verified. Thus, the transgenic 16HBE cell lines were successfully established. Cells expressing oncogene H-Ras or c-Myc grew 30.3% or 10.4% faster than control cells. However, these cells failed to form colonies in soft agar or form tumor in nude mice. 16HBETR, 16HBETM cells obtained transformed phenotype at 5 wks, 11 wks, respectively after treatment with BPDE, which are 15 wks and 9 wks earlier than control cells 16HBETV (20 wks). Meanwhile, 16HBETR, 16HBETM cells obtained transformed phenotype at 11 wks, 14 wks, respectively after treatment with nickel sulfate, which are 21 wks and 18 wks earlier than control cells (32 wks). CONCLUSION: With the advantage of shorter latency, transgenic human cell transformation models could be used in potent carcinogen screening and applied to chemical-carcinogenesis mechanism study.


Cell Line , Cell Transformation, Neoplastic , Epithelial Cells , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Animals , Carcinogenicity Tests , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression , Gene Expression Regulation , Genes, myc , Genes, ras , Humans , Mice , Mice, Inbred BALB C , Mice, Nude
16.
Toxicol Appl Pharmacol ; 232(3): 478-86, 2008 Nov 01.
Article En | MEDLINE | ID: mdl-18778725

To develop human cell models for assessing the carcinogenic potential of chemicals, we established transgenic human cell lines and tested the sensitivity of known carcinogens using a cell transformation assay. A retroviral vector encoding an oncogenic allele of H-Ras (HBER) or c-Myc (HBEM) was introduced into human bronchial epithelial cells (HBE) immortalized by SV40 large T (LT) antigen, leading to increased cell proliferation but failing to confer a transformed phenotype characterized by anchorage-independent cell growth and tumor formation of immunodeficient mice. When these pre-transformed cells were treated with nickel sulfate (NiSO4), we found that it shortened the latency of malignant transformation at least by 19 wk in HBER cells or 16 wk in HBEM cells compared to vector control cells. Similarly, the latency of cell transformation was shorter by 15 wk in HBER cells or 9 wk in HBEM cells when cells were treated with benzo(a)pyrenediol epoxide (BPDE). HBER cells appeared to be more sensitive to TPA, NiSO4 or BPDE-induced cell transformation compared to human embryonic kidney cells expressing H-Ras (HEKR), implying that cell-type specificity is one of important factors determining the effectiveness of the assay. Using AFB1 and BaP as the representative pro-carcinogens, we also compared the efficiency of three different metabolic conditions in mediating cell transformation. Low dose chemical induction seems to be a prospective system used for metabolic activation of pro-carcinogens. Our findings provided direct evidence that a genetically modified human cell transformation model can be applied to the assessment of potent carcinogens.


Bronchi/drug effects , Carcinogenicity Tests/methods , Cell Transformation, Neoplastic , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Aflatoxin B1/pharmacokinetics , Aflatoxin B1/toxicity , Animals , Benzo(a)pyrene/pharmacokinetics , Benzo(a)pyrene/toxicity , Bronchi/metabolism , Cell Line , Chromosome Aberrations , Cytochrome P-450 CYP1A1/biosynthesis , Genes, myc , Genes, ras , Humans , Mice , Mice, Inbred BALB C , Nickel/toxicity
17.
Wei Sheng Yan Jiu ; 35(1): 7-9, 2006 Jan.
Article Zh | MEDLINE | ID: mdl-16598920

OBJECTIVE: To evaluate and screen the specific RNAi fragments which can effectively inhibit Aryl hydrocarbon receptor(AHR) gene mRNA expression in human bronchial epithelial cell line (16HBE). METHODS: AHR mRNA of 16HBE cells transfected 4 different AHR gene interfere sites were determined quantitatively with the quantitative competitive RT-PCR by using self-prepared internal standard as competitive templates, and the RNA interfere effect wasevaluated. RESULTS: AHR mRNA average expression per 40ng total RNA of 16HBE cells transfected 4 different AHR gene interfere fragments were 5.65fg, 14.78fg, 3.14fg and 0.68fg respectively, the average rates of inhibition were 61.6%, -0.5%, 78.6% and 95.4% respectively. CONCLUSION: AHR gene specific effective RNA interfere sequence ware screened by quantitative competitive RT-PCR which could accurately quantify gene mRNA level, and offered condition for studying the gene function of AHR.


Bronchi/cytology , Epithelial Cells/metabolism , RNA, Small Interfering/genetics , Receptors, Aryl Hydrocarbon/genetics , Binding, Competitive , Cell Line , Epithelial Cells/cytology , Humans , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
18.
Ai Zheng ; 23(3): 282-7, 2004 Mar.
Article Zh | MEDLINE | ID: mdl-15025958

BACKGROUND & OBJECTIVE: Carcinoembryonic antigen (CEA) positive cancers are poorly responded to different kinds of treatments. Gene vaccines are promising in research of gene immunotherapy for these tumors. In this study, a CEA gene vaccine with hIL-2 as an immune adjuvant was constructed into a pVAX1 vector for synchronous expression, so as to explore experimentally a new biotherapy strategy against tumors. METHODS: Using reverse transcription polymerase chain reaction (RT-PCR), CEA cDNA was obtained from a large intestine carcinoma tissue; its encoded protein was compared with the CEA presented in GenBank using the protein analysis software. The acquired CEA cDNA fragment was linked to hIL-2 cDNA via an IRES site and cloned into the pVAX1 vector. The recombinant plasmid was estimated by CEA luminometry assay and hIL-2 ELISA measurement respectively. RESULTS: The nucleotide sequences of the target gene fragments of the recombinant plasmid were verified. The acquired CEA sequence is highly homologous with M29540 and M17303 (99.8%) in GenBank; and the PCR sequence of hIL-2 is coincident with the original cDNA (100%) provided. The antigenicity,membrane-spanning segments, signal cleavage sites, secondary structure and 3D structure of the acquired CEA protein were similar to the original proteins of M29540 and M17303 predicted by the protein analysis software. Results showed the recombinant could steadily express CEA antigen and hIL-2 protein synchronously in CHO cells in vitro. CONCLUSION: The CEA cDNA was obtained from the tumor tissue and the CEA gene vaccine with hIL-2 coexpression was constructed successfully. It has provided a possible method for immunotherapy against CEA positive cancers in vivo.


Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/genetics , Interleukin-2/genetics , Vaccines, DNA/therapeutic use , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Immunotherapy , Molecular Sequence Data , Neoplasms/therapy , Plasmids , Reverse Transcriptase Polymerase Chain Reaction
...