Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
EBioMedicine ; 53: 102693, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32114385

ABSTRACT

BACKGROUND: Treatment of Crohn's disease (CD) remains to be a challenge due to limited insights for its pathogenesis. We aimed to determine the role of O-Linked ß-N-acetylglucosamine (O-GlcNAc) in the development of CD and evaluate therapeutic effects of O-GlcNAc inhibitors on CD. METHODS: O-GlcNAc in intestinal epithelial tissues of CD, adherent-invasive Escherichia coli (AIEC) LF82-infected cells and mice was determined by immunoblot and immunohistochemistry. AIEC LF82 and dextran sulfate sodium were administrated into C57BL/6 mice for estabolishing inflammatory bowel disease model and for therapeutic study. FINDINGS: O-GlcNAc was increased in intestinal epithelial tissues of CD patients and AIEC LF82-infected mice. Infection of AIEC LF82 up-regulated the level of UDP-GlcNAc and increased O-GlcNAc in human colon epithelial HCT116 and HT-29 cells. We identified that IKKß and NF-κB were O-Glycosylated in AIEC LF82-treated cells. Mutations of IKKß (S733A) and p65 (T352A) abrogated the O-GlcNAc in IKKß and NF-κB and inhibited AIEC LF82-induced activation of NF-κB. Application of 6-diazO-5-oxO-L-norleucine, an agent that blocks the production of UDP-GlcNAc and inhibits O-GlcNAc, inactivated NF-κB in AIEC LF82-infected cells, enhanced the formation of autophagy, promoted the removal of cell-associated AIEC LF82, alleviated intestinal epithelial inflammation, and improved the survival of the colitis mice. INTERPRETATION: Intestinal inflammation in CD is associated with increased O-GlcNAc modification, which is required for NF-κB activation and suppression of autophagy. Targeting O-GlcNAc could be an effective therapy for inflammatory bowel disease. FUNDING: National Natural Science Foundation of China (Nos. 81573087 and 81772924) and International Cooperation Foundation of Jilin Province (20190701006GH).


Subject(s)
Acetylglucosamine/metabolism , Crohn Disease/metabolism , NF-kappa B/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Autophagy , Female , HCT116 Cells , HT29 Cells , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL
2.
Aging Dis ; 11(1): 44-59, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32010480

ABSTRACT

Many oncogenes are involved in the progression from low-grade squamous intraepithelial lesions (LSILs) to high-grade squamous intraepithelial lesions (HSILs); which greatly increases the risk of cervical cancer (CC). Thus, a reliable biomarker for risk classification of LSILs is urgently needed. The prolyl isomerase Pin1 is overexpressed in many cancers and contributes significantly to tumour initiation and progression. Therefore, it is important to assess the effects of cancer therapies that target Pin1. In our study, we demonstrated that Pin1 may serve as a biomarker for LSIL disease progression and may constitute a novel therapeutic target for CC. We used a the novel Pin1 inhibitor KPT-6566, which is able to covalently bind to Pin1 and selectively target it for degradation. The results of our investigation revealed that the downregulation of Pin1 by shRNA or KPT-6566 inhibited the growth of human cervical cancer cells (CCCs). We also discovered that the use of KPT-6566 is a novel approach to enhance the therapeutic efficacy of cisplatin (DDP) against CCCs in vitro and in vivo. We showed that KPT-6566-mediated inhibition of Pin1 blocked multiple cancer-driving pathways simultaneously in CCCs. Furthermore, targeted Pin1 treatment suppressed the metastasis and invasion of human CCCs, and downregulation of Pin1 reversed the epithelial-mesenchymal transition (EMT) of CCCs via the c-Jun/slug pathway. Collectively, we showed that Pin1 may be a marker for the risk of progression to HSIL and that inhibition of Pin1 has anticancer effects against CC.

3.
Int J Cardiol ; 259: 153-162, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29579593

ABSTRACT

BACKGROUND: The AMP-activated protein kinase (AMPK) plays critical roles in growth regulation and metabolism reprogramming. AMPK activation protects cells against apoptosis from injury in different cell and animal models. However, its function in necroptosis remains largely unclear. METHODS AND RESULTS: In the current study, we demonstrated that AMPK was activated upon necroptosis induction and protected mouse embryonic fibroblasts (MEFs) and cardiomyocytes from N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) and reactive oxygen species (ROS) induced necroptosis. Activation of AMPK with chemicals A-769662, 2-deoxyglucose (2-DG), and metformin or constitutively active (CA) AMPK markedly decreased necroptosis and cytotoxicity induced by MNNG. In contrast, AMPK inhibitor compound C, dominant negative (DN) AMPK, as well as AMPK shRNAs increased necroptosis and cytotoxicity induced by MNNG. We further showed that AMPK physically associated with a protein complex containing PGAM5 and Keap1 whereby facilitating Keap1-mediated PGAM5 ubiquitination upon necroptosis induction. The AMPK agonist metformin ameliorated myocardial ischemia and reperfusion (IR) injury and reduced necroptosis through down-regulating the expression of PGAM5 in the Langendorff-perfused rat hearts. CONCLUSION: Activation of AMPK protects against necroptosis via promoting Keap1-mediated PGAM5 degradation. Metformin may act as a valuable agent for the protection of myocardial ischemia and reperfusion injury by activating AMPK and reducing necroptosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Cell Line , Enzyme Activation/physiology , Isolated Heart Preparation/methods , Male , Mice , Mice, Knockout , Necrosis/metabolism , Necrosis/pathology , Necrosis/prevention & control , Rats , Rats, Sprague-Dawley
4.
Oncotarget ; 9(2): 1885-1897, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29416738

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Mutations of TP53 may reach 70% - 85% in HNSCC patients without human papillomavirus (HPV) infection. Recurrence rate remains particularly high for HNSCC patients with mutations in the TP53 gene although patients are responsive to surgery, irradiation, and chemotherapy early in the treatment. p53-Reactivation and Induction of Massive Apoptosis-1 (PRIMA-1) and its methylated analogue PRIMA-1Met (also known as APR-246) are quinuclidine compounds that rescue the DNA-binding activity of mutant p53 (mut-p53) and restore the potential of wild-type p53. In the current report, we demonstrated that inhibition of poly (ADP-ribose) polymerase-1 (PARP-1) with 6(5H)-phenanthridinone (PHEN) and N-(6-Oxo-5,6-dihydrophenanthridin-2-yl)-(N, N-dimethylamino) acetamide hydrochloride (PJ34) sensitizes UMSCC1, UMSCC14, and UMSCC17A, three HNSCC cell lines to the treatment of APR-246. PHEN enhances APR-246-induced apoptosis, but not programmed necrosis or autophagic cell death in HNSCC cells. The PARP-1 inhibition-induced sensitization of HNSCC cells to APR-246 is independent of TP53 mutation. Instead, PARP-1 inhibition promotes APR-246-facilitated inactivation of thioredoxin reductase 1 (TrxR1), leading to ROS accumulation and DNA damage. Overexpression of TrxR1 or application of antioxidant N-acetyl-L-cysteine (NAC) depletes the ROS increase, reduces DNA damage, and decreases cell death triggered by APR-246/PHEN in HNSCC cells. Thus, we have characterized a new function of PARP-1 inhibitor in HNSCC cells by inactivation of TrxR1 and elevation of ROS and provide a novel therapeutic strategy for HNSCC by the combination of PARP-1 inhibitors and APR-246.

5.
Oncogene ; 37(25): 3384-3398, 2018 06.
Article in English | MEDLINE | ID: mdl-29348462

ABSTRACT

TP53 mutations frequently occur in head and neck squamous cell carcinoma (HNSCC) patients without human papillomavirus infection. The recurrence rate for these patients is distinctly high. It has been actively explored to identify agents that target TP53 mutations and restore wild-type (WT) TP53 activities in HNSCC. PRIMA-1 (p53-reactivation and induction of massive apoptosis-1) and its methylated analogue PRIMA-1Met (also called APR-246) were found to be able to reestablish the DNA-binding activity of p53 mutants and reinstate the functions of WT p53. Herein we report that piperlongumine (PL), an alkaloid isolated from Piper longum L., synergizes with APR-246 to selectively induce apoptosis and autophagic cell death in HNSCC cells, whereas primary and immortalized mouse embryonic fibroblasts and spontaneously immortalized non-tumorigenic human skin keratinocytes (HaCat) are spared from the damage by the co-treatment. Interestingly, PL-sensitized HNSCC cells to APR-246 are TP53 mutation-independent. Instead, we demonstrated that glutathione S-transferase pi 1 (GSTP1), a GST family member that catalyzes the conjugation of GSH with electrophilic compounds to fulfill its detoxification function, is highly expressed in HNSCC tissues. Administration of PL and APR-246 significantly suppresses GSTP1 activity, resulting in the accumulation of ROS, depletion of GSH, elevation of GSSG, and DNA damage. Ectopic expression of GSTP1 or pre-treatment with antioxidant N-acetyl-L-cysteine (NAC) abrogates the ROS elevation and decreases DNA damage, apoptosis, and autophagic cell death prompted by PL/APR-246. In addition, administration of PL and APR-246 impedes UMSCC10A xenograft tumor growth in SCID mice. Taken together, our data suggest that HNSCC cells are selectively sensitive to the combination of PL and APR-246 due to a remarkably synergistic effect of the co-treatment in the induction of ROS by suppression of GSTP1.


Subject(s)
Carcinoma, Squamous Cell/pathology , Dioxolanes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glutathione S-Transferase pi/metabolism , Head and Neck Neoplasms/pathology , Quinuclidines/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Case-Control Studies , Cell Proliferation , Drug Therapy, Combination , Female , Follow-Up Studies , Glutathione S-Transferase pi/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Male , Mice , Mice, SCID , Prognosis , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
6.
Oncotarget ; 7(45): 73389-73401, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27705915

ABSTRACT

Liver kinase B1 (LKB1) functions as a tumor suppressor encoded by STK11, a gene that mutated in Peutz-Jeghers syndrome and in sporadic cancers. Previous studies showed that LKB1 participates in IR- and ROS-induced DNA damage response (DDR). However, the impact of LKB1 mutations on targeted cancer therapy remains unknown. Herein, we demonstrated that LKB1 formed DNA damage-induced nuclear foci and co-localized with ataxia telangiectasia mutated kinase (ATM), γ-H2AX, and breast cancer susceptibility 1 (BRCA1). ATM mediated LKB1 phosphorylation at Thr 363 following the exposure of cells to ionizing radiation (IR). LKB1 interacted with BRCA1, a downstream effector in DDR that is recruited to sites of DNA damage and functions directly in homologous recombination (HR) DNA repair. LKB1 deficient cells exhibited delayed DNA repair due to insufficient HR. Notably, LKB1 deficiency sensitized cells to poly (ADP-ribose) polymerase (PARP) inhibitors. Thus, we have demonstrated a novel function of LKB1 in DNA damage response. Cancer cells lacking LKB1 are more susceptible to DNA damage-based therapy and, in particular, to drugs that further impair DNA repair, such as PARP inhibitors.


Subject(s)
DNA Damage , Drug Resistance, Neoplasm , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/metabolism , Cell Line, Tumor , DNA Repair , Homologous Recombination , Humans , Phosphorylation , Protein Binding , Protein Transport
7.
Proc Natl Acad Sci U S A ; 113(33): 9333-8, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27482104

ABSTRACT

High-risk human papillomaviruses (HPVs) are causative agents of anogenital cancers and a fraction of head and neck cancers. The mechanisms involved in the progression of HPV neoplasias to cancers remain largely unknown. Here, we report that O-linked GlcNAcylation (O-GlcNAc) and O-GlcNAc transferase (OGT) were markedly increased in HPV-caused cervical neoplasms relative to normal cervix, whereas O-GlcNAcase (OGA) levels were not altered. Transduction of HPV16 oncogene E6 or E6/E7 into mouse embryonic fibroblasts (MEFs) up-regulated OGT mRNA and protein, elevated the level of O-GlcNAc, and promoted cell proliferation while reducing cellular senescence. Conversely, in HPV-18-transformed HeLa cervical carcinoma cells, inhibition of O-GlcNAc with a low concentration of a chemical inhibitor impaired the transformed phenotypes in vitro. We showed that E6 elevated c-MYC via increased protein stability attributable to O-GlcNAcylation on Thr58. Reduction of HPV-mediated cell viability by a high concentration of O-GlcNAc inhibitor was partially rescued by elevated c-MYC. Finally, knockdown of OGT or O-GlcNAc inhibition in HeLa cells or in TC-1 cells, a mouse cell line transformed by HPV16 E6/E7 and activated K-RAS, reduced c-MYC and suppressed tumorigenesis and metastasis. Thus, we have uncovered a mechanism for HPV oncoprotein-mediated transformation. These findings may eventually aid in the development of effective therapeutics for HPV-associated malignancies by targeting aberrant O-GlcNAc.


Subject(s)
Carcinogenesis , N-Acetylglucosaminyltransferases/physiology , Oncogene Proteins, Viral/physiology , Repressor Proteins/physiology , Uterine Cervical Neoplasms/etiology , Animals , Cell Line, Tumor , Female , Genes, myc , Humans , Mice , Mice, Inbred C57BL , Papillomavirus E7 Proteins/physiology , Uterine Cervical Neoplasms/virology
8.
Biochem Biophys Res Commun ; 447(2): 224-30, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24709078

ABSTRACT

Antitumor antibiotic lidamycin (LDM) is widely used in the treatment of a variety of cancers. Here we demonstrated that LDM up-regulates the expression of the tumor suppressor p53 gene by repressing Oct4 transcription. We showed that low dose LDM-induced increase of p53 expression and decrease of Oct4 expression in P19 and HCT116-p53(+/+) cells. Knockdown of Oct4 expression by siRNA led to activation of p53 in both cell lines, whereas ectopical expression of Oct4 significantly inhibited p53 expression in P19 cells. LDM-induced p53 activation was blocked by ectopical expression of Oct4.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Enediynes/pharmacology , Gene Expression/drug effects , Octamer Transcription Factor-3/genetics , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Mice , Promoter Regions, Genetic/drug effects , RNA, Small Interfering/genetics , Up-Regulation
9.
Chin Med J (Engl) ; 126(18): 3539-44, 2013.
Article in English | MEDLINE | ID: mdl-24034105

ABSTRACT

BACKGROUND: Claudin-6 is a protein component of tight junctions and its expression could downregulate the malignant phenotype of breast carcinoma. Here we investigated the mechanisms of claudin-6 induced human MCF-7 breast cancer cells apoptosis, invasion, and migration. METHODS: Terminal deoxyribonucleotide transferase-mediated nick-end labeling assay and Annexin-V/PI double stain assay were carried out to evaluate apoptosis. Inhibitors of each pathway were used to inactivate the signaling pathways. The expression of claudin-6 and phosphate p38, Erk 1/2 and Akt protein levels was confirmed by Western blotting analysis. Invasive and migratory traits of claudin-6 expressing cells were determined by Boyden chamber invasion assay and monolayer wound-healing assay. RESULTS: Cells with high-level expression of claudin-6 had a higher rate of apoptosis than control cells. Western blotting assay showed that by contrast to control groups, p38 pathways were more activated in claudin-6 expressing cells. However, after inhibitor SB203580 treatment, the activation status could be significantly counteracted. Furthermore, by applying inhibitors to the apoptotic rate, invasive and migratory traits were also recovered in cells with claudin-6 expression. CONCLUSION: Claudin-6 may function through p38 mitogen-activated protein kinase pathway, of which inhibition may reverse claudin-6-induced cell apoptosis, invasion, and migration.


Subject(s)
Apoptosis/physiology , Cell Movement/physiology , Claudins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis/genetics , Blotting, Western , Cell Movement/genetics , Claudins/genetics , Humans , In Situ Nick-End Labeling , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , MCF-7 Cells , p38 Mitogen-Activated Protein Kinases/genetics
10.
Anat Rec (Hoboken) ; 296(4): 595-603, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23408563

ABSTRACT

Cisplatin resistance is a difficult problem in clinical chemotherapy, and the mechanisms involved in cisplatin resistance require further study. In this study, we investigated the role of chloride channel-3 (ClC-3) in cisplatin resistance. Autophagy was demonstrated by accumulation of LC3-II, beclin 1 and Atg12-Atg5. The ultrastructure changes were observed under electron microscope. Chemical staining with acridine orange or MDC was used to detect acidic vesicular organelles. Quantification of apoptosis was detected by PI and Annexin V staining. The mechanisms involved in the Akt pathway and autophagy were studied by western blot analysis. Our results showed that Akt phosphorylation and autophagy were induced by cisplatin in human glioma U251 cells. Specific inhibition of ClC-3 by ClC-3 siRNA sensitized the apoptosis-resistant U251 cells to cisplatin-mediated cell death and downregulated phosphorylated Akt. Interestingly, ClC-3 suppression also inhibited induction of autophagy by cisplatin although the Akt/mTOR pathway was deregulated. Counteracting the autophagic process by 3-methylademine enhanced cytotoxicity of cisplatin, revealing that autophagy plays a key role in chemoresistance. Suppressing the Akt/mTOR pathway by the NADPH oxidase inhibitor diphenyl iodonium (DPI) indicated that cisplatin-induced activation of Akt/mTOR pathway requires generation of reactive oxygen species (ROS) through NADPH oxidase. Collectively, our results suggest that ClC-3 suppression causes the inhibition of Akt and autophagy, which can enhance the therapeutic benefit of cisplatin in U251 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Brain Neoplasms/enzymology , Chloride Channels/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Glioma/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/ultrastructure , Cell Line, Tumor , Chloride Channels/genetics , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glioma/genetics , Glioma/ultrastructure , Humans , Microscopy, Electron, Transmission , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Phosphorylation , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Staining and Labeling , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection
11.
Zhonghua Bing Li Xue Za Zhi ; 39(1): 44-7, 2010 Jan.
Article in Chinese | MEDLINE | ID: mdl-20388399

ABSTRACT

OBJECTIVE: To explore the role of estrogen in the regulation of the expression of claudin-6 and biological behavior in MCF-7 cells. METHODS: RT-PCR and immunocytochemistry were conducted to analyze the expression and localization of claudin-6 in MCF-7 cells treated with 17ß-estradiol. CCK-8 kit assay and Scratch Test were conducted to analyze the capability of proliferation and migration of 17ß-estradiol treated MCF-7 cells. RESULTS: RT-PCR analysis and immunocytochemistry showed that 17ß-estradiol induced a concentration-and time-dependent effect on claudin-6. At 5 nmol/L and at 24 h, 17ß-estradiol treatment led to an increased level of claudin-6, which was located in the membrane of MCF-7 cells. CCK-8 analysis showed a significant decrease in the capability of proliferation of MCF-7 cells compared with the control group (P < 0.05). Cells Scratch Test showed decreased migration capability of MCF-7 cells compared with the control group (P > 0.05). CONCLUSIONS: 17ß-E2 might regulate the expression of claudin-6 and inhibit the proliferation and migration of MCF-7 cells. The inhibitory effects of 17ß-E2 on growth and migration of MCF-7 cells may be mediated by claudin-6 expression regulation.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Estradiol/pharmacology , Membrane Proteins/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Claudins , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Female , Humans
12.
Zhonghua Yi Xue Za Zhi ; 90(6): 407-12, 2010 Feb 09.
Article in Chinese | MEDLINE | ID: mdl-20367941

ABSTRACT

OBJECTIVE: To explore the effect of over-expression of tight junction protein claudin-6 upon the biological characters of breast cancer cell line MCF-7. METHODS: The MCF-7 subline expressing a high level of claudin-6 was established by transfection with a pcDNA3.1-claudin-6 expression vector. The expression of claudin-6 in mRNA and its protein level were confirmed by RT-PCR, Western blot and immunofluorescent assays. Then the effect of claudin-6 upon cell proliferation was examined by MTT assay. Colony-forming assays were used to examine 2-D and 3-D colony-forming capacities. Invasive and migratory traits of claudin-6 expressing cells were determined by Boyden chamber invasion assay and monolayer wound-healing assay. The structure and function of tight junctions in both parental and claudin-6 expression MCF-7 cells were evaluated by measuring transepithelial electrical resistance. RESULTS: Immunofluorescent assays showed that transfected cells expressed claudin-6 on their membrane. Cells with a high level expression of claudin-6 grew slowly than control cells. Anchorage-independent growth, invasive and migratory traits also decreased substantially in cells with claudin-6 expression; whereas the transepithelial electrical resistance increased in the claudin-6 transfected cells. CONCLUSION: The up-regulation of claudin-6 expression in MCF-7 breast cancer cells suppresses their malignant phenotypes with a correlation with the restoration of tight junction integrity. Claudin-6 may function as a cancer suppressor whose down-regulation contributes to the malignant progression of certain types of breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Membrane Proteins/metabolism , Tight Junctions/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Claudins , Down-Regulation , Female , Humans , Neoplasm Invasiveness , Tight Junctions/pathology , Transfection
13.
Zhonghua Er Bi Yan Hou Ke Za Zhi ; 39(3): 157-61, 2004 Mar.
Article in Chinese | MEDLINE | ID: mdl-15283295

ABSTRACT

OBJECTIVE: To detect the expression of bcl-2 and bax in each phase of cell cycle in laryngeal carcinoma, and explore the relationship between the expression of bcl-2 and bax in each phase of the cell cycle and the occurrence, development and prognosis in the carcinoma of larynx. METHODS: The immunohistochemical method, TUNEL technique and flowcytometry (FCM) parameter analyses were combined to detect the apoptosis and the expression of bcl-2 and bax in each phase of the cell cycle in 15 polyps of vocal cord and 387 laryngeal carcinomas. RESULTS: Total bcl-2 expression and bcl-2 expression in G0G1 stage in laryngeal carcinoma was significantly higher than that in polyp of vocal cord. In contrast, the total bax expression and the bax expression in each phase of cell cycle in laryngeal carcinoma were all lower than that in polyp of vocal cord. The total apoptosis index in laryngeal carcinoma was obviously lower than that in polyp of vocal cord, and this phenomenon was mainly caused by the decrease of the apoptosis in G0G1 phase. The bcl-2, bax expression and the apoptosis wasn't notably related to clinical stage, clinical type and T grade. In poor-differentiated squamous carcinoma, the bcl-2 expression in S and G2M phase was obviously higher than that in well-differentiated and the moderate-differentiated squamous carcinoma. The total apoptosis index, the apoptosis in S phase and the apoptosis in G2M phase were obviously enhanced both in the group of recurrence and in the patients who died in 5 years after the operation, in the same samples, the significant increasing of bcl-2 expression in S and G2M phase was detected. CONCLUSIONS: The significant decreasing of the apoptosis in G0G1 phase caused by high expression of bcl-2 was an important affair in the initial stage of laryngeal carcinoma. Accompanying the significant increasing of the total apoptosis index, the apoptosis in S phase and the apoptosis in G2M phase could be regard as an indicator that the cancer of larynx was malignant with poor prognosis and need adjuvant therapy. The decreasing of bax expression may play a role in the occurrence of laryngeal carcinoma.


Subject(s)
Carcinoma, Squamous Cell/genetics , Genes, bcl-2 , Laryngeal Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Adult , Aged , Apoptosis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Female , Gene Expression Regulation, Developmental , Humans , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Male , Middle Aged , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...