1
|
WANG J, JIN QG, LIU RP, WANG XQ, LI YH, KIM NH, XU YN. Dihydromyricetin supplementation during in vitro culture improves porcine oocyte developmental competence by regulating oxidative stress. J Reprod Dev 2023; 69:10-17. [PMID: 36403957 PMCID: PMC9939282 DOI: 10.1262/jrd.2022-031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dihydromyricetin (DHM), a dihydroflavonoid compound, exhibits a variety of biological activities, including antitumor activity. However, the effects of DHM on mammalian reproductive processes, especially during early embryonic development, remain unclear. In this study, we added DHM to porcine zygotic medium to explore the influence and underlying mechanisms of DHM on the developmental competence of parthenogenetically activated porcine embryos. Supplementation with 5 μM DHM during in vitro culture (IVC) significantly improved blastocyst formation rate and increased the total number of cells in porcine embryos. Further, DHM supplementation also improved glutathione levels and mitochondrial membrane potential; reduced natural reactive oxygen species levels in blastomeres and apoptosis rate; upregulated Nanog, Oct4, SOD1, SOD2, Sirt1, and Bcl2 expression; and downregulated Beclin1, ATG12, and Bax expression. Collectively, DHM supplementation regulated oxidative stress during IVC and could act as a potential antioxidant during in vitro porcine oocytes maturation.
Collapse
Affiliation(s)
- Jing WANG
- College of Agriculture, Yanbian University, Yanji 133000, China,Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529000,
China
| | - Qing-Guo JIN
- College of Agriculture, Yanbian University, Yanji 133000, China
| | - Rong-Ping LIU
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529000,
China
| | - Xin-Qin WANG
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529000,
China
| | - Ying-Hua LI
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529000,
China
| | - Nam-Hyung KIM
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529000,
China
| | - Yong-Nan XU
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529000,
China
| |
Collapse
|
2
|
Mehrzadi S, Pourhanifeh MH, Mirzaei A, Moradian F, Hosseinzadeh A. An updated review of mechanistic potentials of melatonin against cancer: pivotal roles in angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress. Cancer Cell Int 2021; 21:188. [PMID: 33789681 PMCID: PMC8011077 DOI: 10.1186/s12935-021-01892-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/23/2021] [Indexed: 12/19/2022] Open
Abstract
Cancers are serious life-threatening diseases which annually are responsible for millions of deaths across the world. Despite many developments in therapeutic approaches for affected individuals, the rate of morbidity and mortality is high. The survival rate and life quality of cancer patients is still low. In addition, the poor prognosis of patients and side effects of the present treatments underscores that finding novel and effective complementary and alternative therapies is a critical issue. Melatonin is a powerful anticancer agent and its efficiency has been widely documented up to now. Melatonin applies its anticancer abilities through affecting various mechanisms including angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress. Regarding the implication of mentioned cellular processes in cancer pathogenesis, we aimed to further evaluate the anticancer effects of melatonin via these mechanisms.
Collapse
Affiliation(s)
- Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mirzaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Farid Moradian
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Qian B, Yao Z, Yang Y, Li N, Wang Q. Downregulation of SDCBP inhibits cell proliferation and induces apoptosis by regulating PI3K/AKT/mTOR pathway in gastric carcinoma. Biotechnol Appl Biochem 2021; 69:240-247. [PMID: 33432665 DOI: 10.1002/bab.2103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/06/2021] [Indexed: 01/04/2023]
Abstract
Syndecan-binding protein (SDCBP) has been reported to critically process a core role in tumorigenesis. This study was conducted to characterize a novel regulatory network of SDCBP in gastric carcinoma (GC) cells. Our findings indicated that overexpression of SDCBP promoted the proliferation of GC cell and increased proliferating cell nuclear antigen (PCNA) expression. Moreover, the overexpression of SDCBP suppressed the apoptosis of GC cell along with a decrease of Bax/Bcl-2 ratio and induction of PI3K/AKT/mTOR activation. However, knockdown of SDCBP exhibited opposed effects on GC cells. Furthermore, silencing SDCBP significantly inhibited GC cell viability and PCNA expression accompanied with the upregulated cell apoptosis and Bax/Bcl-2 ratio, which was regulated by PI3K/AKT/mTOR signaling pathway. And it was further determined that PI3K inhibitor LY294002, AKT inhibitor Torin1, and mTOR inhibitor MK-2206 suppressed the apoptosis. In conclusion, SDCBP promotes the growth ability of GC by inducing the PCNA expression and inhibiting GC cell apoptosis via inactivation of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Bo Qian
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Zhiheng Yao
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yang Yang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Qiao Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
4
|
Yang XF, Zhou SY, Wang C, Huang W, Li N, He F, Li FR. Inhibition of LSD1 promotes the differentiation of human induced pluripotent stem cells into insulin-producing cells. Stem Cell Res Ther 2020; 11:185. [PMID: 32430053 PMCID: PMC7238565 DOI: 10.1186/s13287-020-01694-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human induced pluripotent stem cells (hiPSCs) represent a potentially unlimited source of pancreatic endocrine lineage cells. Although insulin-producing β cells derived from hiPSCs have been successfully induced, much work remains to be done to achieve mature β cells. Lysine-specific demethylase 1 (LSD1) plays an important role in the regulation of hiPSC self-renewal and differentiation. We propose a new strategy to acquire insulin-producing cells (IPCs) from hiPSCs by knocking down LSD1. METHODS Knockdown of LSD1 in hiPSCs with five shRNA. Assessment of the effects of shRNA on hiPSC proliferation, cell cycle, and apoptosis. Using knockdown hiPSCs with 31.33% LSD1 activity, we achieved a four-step differentiation into IPCs and test its differentiation efficiency, morphology, and marker genes and proteins. We implanted the IPCs into the renal subcapsular of SCID-Beige diabetic mice to evaluate the hypoglycemic effect in vivo. We tested LSD1 and HDAC1 whether they are present in the CoREST complex through IP-WB, and analyzed LSD1, CoREST, HDAC1, H3K4me2/me3, and H3K27me3 protein expression before and after knockdown of LSD1. RESULTS Differentiated hiPSCs were 38.32% ± 3.54% insulin-positive cells and released insulin/C-peptide in response to glucose stimulus in a manner comparable to adult human islets. Most of the IPCs co-expressed mature β cell-specific markers. When transplanted under the left renal capsule of SCID-Beige diabetic mice, these IPCs reversed hyperglycemia, leading to a significant increase in the definitive endoderm cells. IP-WB results showed that LSD1, HDAC1, and CoREST formed a complex in hiPSCs. Chip-PCR results showed that LSD1, HDAC1, and CoREST were enriched in the same district during the SOX17 and FOXA2 promoter region. Inhibition of LSD1 would not affect the level of CoREST but decreased the HDAC1 expressions. The H3K4me2/me3 and H3K9act level of SOX17 and FOXA2 promoter region increased after inhibited of LSD1, and promoted transcriptional activation. The H3K4me2/me3 and H3K9act level of OCT4 and SOX2 promoter region decreased with the transcriptional repressed. CONCLUSIONS LSD1 regulated histone methylation and acetylation in promoter regions of pluripotent or endodermal genes. Our results suggest a highly efficient approach to producing IPCs from hiPSCs.
Collapse
Affiliation(s)
- Xiao-Fei Yang
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China.,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Shu-Yan Zhou
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China
| | - Ce Wang
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Wei Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ning Li
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China
| | - Fei He
- Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China.,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Fu-Rong Li
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China. .,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China. .,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China.
| |
Collapse
|
5
|
Zhang Z, Li C, Shang L, Zhang Y, Zou R, Zhan Y, Bi B. Sulforaphane induces apoptosis and inhibits invasion in U251MG glioblastoma cells. SPRINGERPLUS 2016; 5:235. [PMID: 27026929 PMCID: PMC4771656 DOI: 10.1186/s40064-016-1910-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
In recent studies, sulforaphane (SFN) has been seen to demonstrate antioxidant and anti-tumor activities. In the present study, the viability inhibition effects of SFN in U251MG glioblastoma cells were analyzed by MTS. Morphology changes were observed by microscope. Apoptotic effects of SFN were evaluated by annexin V binding capacity with flow cytometric analysis. Invasion inhibition effects of SFN were tested by the invasion assay. The molecular mechanisms of apoptotic effects and invasion inhibition effects of SFN were detected by western blot and gelatin zymography. The results indicated that SFN has potent apoptotic effects and invasion inhibition effects against U251MG glioblastoma cells. These effects are both dose dependent. Taken together, SFN possessed apoptotic activity on U251MG cells indicated by increased annexin V-binding capacity, Bad, Bax, cytochrome C expression, and decreased Bcl-2 and survivin expressions. SFN inhibited invasion in U251MG cells via upregulation of E-cadherin and downregulation of MMP-2, MMP-9 and Galectin-3.
Collapse
Affiliation(s)
- Zhen Zhang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | - Chunliu Li
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | - Li Shang
- Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | | | - Rong Zou
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | - Yan Zhan
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | | |
Collapse
|
6
|
Chen D, Zheng X, Kang D, Yan B, Liu X, Gao Y, Zhang K. Apoptosis and expression of the Bcl-2 family of proteins and P53 in human pancreatic ductal adenocarcinoma. Med Princ Pract 2012; 21:68-73. [PMID: 22024503 DOI: 10.1159/000332423] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/12/2011] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The purpose of this study was to clarify the association between P53 and the Bcl-2 family (Bcl-2, Bax, Bcl-xL, Bcl-xS) expression and apoptosis in pancreatic ductal adenocarcinoma (PDAC). SUBJECTS AND METHODS A total of 70 patients with PDAC were studied. The expression of P53 protein in PDAC was assessed using the immunohistochemical method, which categorized the PDAC patients into two groups: group 1: 36 cases with immunonegative P53(-), and group 2: 34 cases with immunopositive P53(+). The expression of Bcl-2, Bax, Bcl-xL, and Bcl-xS in the 70 PDAC cases was detected by immunohistochemical and Western blotting methods. The apoptotic index (AI) was also measured in these samples by the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) method. The relation between P53 and the Bcl-2 protein family and apoptosis was then evaluated. RESULTS Bcl-2 and Bcl-xS expression was significantly associated with P53 (p < 0.05). No clear associations were found among P53, Bax and Bcl-xL expression (p > 0.05). The AI of groups 1 and 2 was 12.1 ± 2.47 and 8.1 ± 1.48, respectively (p = 0.023). There was no relationship between AI and Bcl-2, Bax, Bcl-xL and Bcl-xS expression (p > 0.05, respectively). Bcl-2/Bax ratio was significantly associated with AI (p < 0.01). CONCLUSION Bcl-2 and Bcl-xS represent significant anti- and proapoptotic proteins, respectively, modulated through a P53-dependent pathway in PDAC, and P53 modulated apoptosis mainly through Bcl-2/Bax ratio.
Collapse
Affiliation(s)
- Dong Chen
- Department of General Surgery, Affiliated Hospital of Medical College, QingDao University, QingDao, ShanDong Province, People’s Republic of China
| | | | | | | | | | | | | |
Collapse
|
7
|
Zhang YJ, Dai Q, Wu SM, Zhu HY, Shen GF, Li EL, Xiao SD. Susceptibility for NSAIDs-induced apoptosis correlates to p53 gene status in gastric cancer cells. Cancer Invest 2008; 26:868-77. [PMID: 18798056 DOI: 10.1080/07357900801944872] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The anti-tumor effect of non-steroidal anti-inflammatory drugs (NSAIDs) remains unclear. Here, we found that the susceptibility for NSAIDs-induced apoptosis might correlate with the status of the p53 gene in gastric cancer cells. Apoptosis in gastric cancer cells expressing wild-type p53 is induced through up-regulation of bax and down-regulation of bcl-2 and that regulation of the bax-bcl-2 heterodimer may be a major target of NSAIDs. As to gastric cancer cells expressing mutant-type p53, other key factors may exist in the NSAIDs' growth inhibition action.
Collapse
Affiliation(s)
- Yan-Jie Zhang
- Digestive Department of the No. 3 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
8
|
Wincewicz A, Sulkowska M, Koda M, Sulkowski S. Cumulative expression of HIF-1-alpha, Bax, Bcl-xL and P53 in human colorectal cancer. Pathology 2007; 39:334-8. [PMID: 17558861 DOI: 10.1080/00313020701329765] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIMS AND METHODS Hypoxia-inducible factor (HIF-1) which contains oxygen regulated HIF-1alpha subunit maintains cytoprotective defence against hypoxic injury by induction of numerous genes. However, apoptotic regulators such as Bcl-xL, Bax and P53 have not been associated with HIF-1 dependent regulation in immunohistochemical evaluation of human colorectal cancer tumours so far. Thus, we visualised these proteins immunohistochemically and using Spearman's test compared for the first time their expression in regard to different clinicopathological traits in 123 (113 for P53 evaluation) human colorectal cancers. RESULTS HIF-1alpha correlated with Bcl-xL or Bax in all patients and particularly in node negative and node positive cancers, deeper intramural tumours (pT3+pT4) and adenocarcinomas. There was no significance in a small group of tumours with lesser extent through intestinal walls (pT1+pT2). In addition HIF-1alpha associated with Bcl-xL in mucinous cancers. Moreover, HIF-1alpha correlated with Bcl-xL or Bax in moderately (G2) and poorly differentiated (G3) cancers, rectal and colonic tumours and in different sex and age groups. P53 correlated only with Bax exclusively in younger patients. CONCLUSIONS HIF-1alpha may influence expression of Bax or Bcl-xL, at least indirectly, as correlations between HIF-1alpha and Bax or Bcl-xL occur constantly.
Collapse
Affiliation(s)
- Andrzei Wincewicz
- Department of Pathology, Medical University of Bialystok, Bialystok, Poland.
| | | | | | | |
Collapse
|
9
|
Zhang M, Ling Y, Yang CY, Liu H, Wang R, Wu X, Ding K, Zhu F, Griffith BN, Mohammad RM, Wang S, Yang D. A novel Bcl-2 small molecule inhibitor 4-(3-methoxy-phenylsulfannyl)-7-nitro-benzofurazan-3-oxide (MNB)-induced apoptosis in leukemia cells. Ann Hematol 2007; 86:471-81. [PMID: 17396262 DOI: 10.1007/s00277-007-0288-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Accepted: 03/12/2007] [Indexed: 10/23/2022]
Abstract
A novel small molecule inhibitor, 4-(3-methoxy-phenylsulfannyl)-7-nitro-benzofurazan-3-oxide (MNB), competes with the Bak BH3 peptide to bind Bcl-2 protein with a binding affinity of IC(50) = 0.70 microM, as assessed by a fluorescence polarization based binding assay. HL-60 cells express the highest levels of Bcl-2 among the cell lines examined. Treated with 5 microM of MNB only for 6 h, 85% of HL-60 cells were detected to undergo apoptosis. Pan-caspase inhibitor, Z-VAD-FMK, blocks MNB-induced apoptosis in HL-60 cells. Caspase-2, caspase-3, caspase-7, caspase-8, caspase-9, and PARP activation were observed at as early as 4 to 6 h of MNB treatment. In addition, it has been confirmed that the caspase-3 specific inhibitor, Z-DEVD-FMK, blocks the activation of caspase-8 in MNB-treated HL-60 cells. MNB treatment does not change Bcl-2 or Bax expression level in HL-60 cells, but causes Bid cleavage. Further experiments have illustrated that MNB inhibits the heterodimerization of Bcl-2 with Bax or Bid, reduces the mitochondrial membrane potential (DeltaPsimt), and induces cytochrome c release from mitochondria in HL-60 cells. These results suggest that MNB induces apoptosis in HL-60 by inhibiting the heterodimerization of Bcl-2 with pro-apoptosis Bcl-2 members, resulting in a decrease in the mitochondrial membrane potential and cytochrome c release, activation of caspases and PARP; it is a caspase-dependent process in which the activation of caspase-8 is dependent on the mitochondrial apoptosis signal transduction pathway. MNB prolongs the life spans of HL-60 bearing mice, potently kills fresh AML and ALL cells, indicating that it has the potential to be developed to treat leukemia.
Collapse
Affiliation(s)
- Manchao Zhang
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wincewicz A, Sulkowska M, Koda M, Kanczuga-Koda L, Witkowska E, Sulkowski S. Significant Coexpression of GLUT-1, Bcl-xL, and Bax in Colorectal Cancer. Ann N Y Acad Sci 2007; 1095:53-61. [PMID: 17404017 DOI: 10.1196/annals.1397.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Hypoxic cancer cells overexpress Glucose transporter 1 (GLUT-1) to accelerate glucose intake mainly for low effective, anaerobic respiration, so that they would not die of oxygen deficiency. Ischemic cell injury triggers apoptosis. Regulators of cell suicide like Bax and Bcl-xL combine their functions to cause apoptosis or to rescue cells from death. GLUT-1, Bax, and Bcl-xL are of prognostic significance in colorectal cancer but they have not been compared, yet. Thus, we aimed to determine eventual correlations between GLUT-1, Bax, and Bcl-xL in association with different clinicopathological features of colorectal cancer patients. Expressions of the proteins were evaluated in specimens of 150 colorectal patients by immunohistochemistry. The levels of tissue expressions were statistically analyzed with Spearman's correlation test. As in group of all the patients, GLUT-1 matched Bcl-xL and Bax in statistically significant manner regardless of different node status, grade of histological differentiation, histopathological type, tumor site, gender and age of patients. GLUT-1 correlated highly with Bcl-xL in both groups of various tumor growth extent: pT1 + pT2 and pT3 + pT4 tumors (P < 0.016, r = 0.6340, P < 0.0001, r = 0.5204, respectively). Bax correlated with GLUT-1 (P < 0.0001, r = 0.4284) and Bcl-xL (P < 0.0001, r = 0.5233) in pT3 and pT4 tumors without any statistical significance in a homologous comparison at pT1 and pT2 stage (P > 0.173, r = 0.1078, P > 0.744, r = 0.1, respectively). Significant coexpression of GLUT-1, Bcl-xL, and Bax could point to cooperation of these regulatory proteins in elimination due to irreversible injury, adaptation to hypoxia, reduction of further damage, and survival of colorectal cancer cells.
Collapse
Affiliation(s)
- Andrzej Wincewicz
- Departments of General and Clinical Pathomorphology, Medical University of Bialystok, Waszyngtona St. 13, 15-269 Bialystok, Poland
| | | | | | | | | | | |
Collapse
|
11
|
Thomadaki H, Scorilas A. BCL2 family of apoptosis-related genes: functions and clinical implications in cancer. Crit Rev Clin Lab Sci 2006; 43:1-67. [PMID: 16531274 DOI: 10.1080/10408360500295626] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
One of the most effective ways to combat different types of cancer is through early diagnosis and administration of effective treatment, followed by efficient monitoring that will allow physicians to detect relapsing disease and treat it at the earliest possible time. Apoptosis, a normal physiological form of cell death, is critically involved in the regulation of cellular homeostasis. Dysregulation of programmed cell death mechanisms plays an important role in the pathogenesis and progression of cancer as well as in the responses of tumours to therapeutic interventions. Many members of the BCL2 (B-cell CLL/lymphoma 2; Bcl-2) family of apoptosis-related genes have been found to be differentially expressed in various malignancies, and some are useful prognostic cancer biomarkers. We have recently cloned a new member of this family, BCL2L12, which was found to be differentially expressed in many tumours. Most of the BCL2 family genes have been found to play a central regulatory role in apoptosis induction. Results have made it clear that a number of coordinating alterations in the BCL2 family of genes must occur to inhibit apoptosis and provoke carcinogenesis in a wide variety of cancers. However, more research is required to increase our understanding of the extent to which and the mechanisms by which they are involved in cancer development, providing the basis for earlier and more accurate cancer diagnosis, prognosis and therapeutic intervention that targets the apoptosis pathways. In the present review, we describe current knowledge of the function and molecular characteristics of a series of classic but also newly discovered genes of the BCL2 family as well as their implications in cancer development, prognosis and treatment.
Collapse
Affiliation(s)
- Hellinida Thomadaki
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Athens, Panepistimiopolis, 15701 Athens, Greece
| | | |
Collapse
|
12
|
Biswas G, Guha M, Avadhani NG. Mitochondria-to-nucleus stress signaling in mammalian cells: nature of nuclear gene targets, transcription regulation, and induced resistance to apoptosis. Gene 2005; 354:132-9. [PMID: 15978749 PMCID: PMC3800739 DOI: 10.1016/j.gene.2005.03.028] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Accepted: 03/23/2005] [Indexed: 12/11/2022]
Abstract
Depletion of mitochondrial DNA (mtDNA) or treatment with mitochondrial poison CCCP initiates mitochondrial stress signaling, which operates through altered Ca2+ homeostasis. In C2C12 rhabdomyoblasts and A549 human lung carcinoma cells mitochondrial stress signaling activates calcineurin and a number of Ca2+ responsive factors including ATF, NFAT, CEBP/delta and CREB. Additionally, PKC and MAP kinase are also activated. A number of nuclear gene targets including those involved in Ca2+ storage/release (RyR1, calreticulin, calsequestrin), glucose metabolism (hexokinase, pyruvate kinase, Glut4), oncogenesis (TGFbeta1, cathepsin L, IGFR1, melanoma antigen) and apoptosis (Bcl-2, Bid, Bad, p53) are upregulated. Mitochondrial stress in both C2C12 myoblasts and A549 cells induced morphological changes and invasive phenotypes. These cells also showed markedly increased resistance to etoposide-induced apoptosis that is a hallmark of highly invasive tumors. Our results describe a new mechanism of altered nuclear gene expression and phenotypic changes triggered by mitochondrial dysfunction and mtDNA damage.
Collapse
Affiliation(s)
| | | | - Narayan G. Avadhani
- Corresponding author. Tel.: +1 215 898 8819; fax: +1 215 573 6651. (N.G. Avadhani)
| |
Collapse
|
13
|
Wang WY, Tang HW, Zhang ZP, Han B, Gao YH, Ma FC, Yan QG, Zhao YL. Expression of Bag-1 and Bcl-2 and its significance in gastric cancer. Shijie Huaren Xiaohua Zazhi 2005; 13:1186-1189. [DOI: 10.11569/wcjd.v13.i10.1186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of Bag-1 and Bcl-2 protein and its clinical significance in gastric cancer (GC).
METHODS: Immunohistochemistry was used to detect the expressison of Bag-1 and Bcl-2 protein in 92 patients with gastric cancer and 40 normal tissues as control.
RESULTS: Compared with normal mucosa, Bag-1 and Bcl-2 were over-expressed in gastric cancerous tissues (60.9% vs 7.5%, 72.8% vs 10%, P<0.01). The expression of Bcl-2 protein was significantly correlated with the differentiation degree (rs = 0.513, P<0.05) and the expression of Bag-1 was significantly associated with Bcl-2 level in gastric cancer (rs = 0.522, P<0.01).
CONCLUSION: The over-expression of Bcl-2 leads to uncontrolled growth and unceasingly increases of GC cells by accelerating cell transformation and proliferation. Bag-1 promotes the function of Bcl-2 and co-expression of Bag-1 and Bcl-2 may mean poor prognosis of GC patients.
Collapse
|
14
|
Yan QG, Shi JG, Huang GS, Zhang CS, Li Q, Hu PZ, Wang WL. In situ quantitative study of BAG-1 and BAD in extrahepatic cholangiocarcinoma. Shijie Huaren Xiaohua Zazhi 2003; 11:1144-1147. [DOI: 10.11569/wcjd.v11.i8.1144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To detect the expression of apoptosis related proteins BAG-1 and BAD and to investigate their functions of apoptosis modulation and relations to tumor differentiation in extrahepatic cholangiocarcinoma.
METHODS Quantitative in situ methods of DAKO EnvisionTM Systems immunohistochemistry in combination with computer-assisted image analysis and apoptotic cells counting were used to observe the expression levels of BAG-1 and BAD and the apoptosis indexes in 42 cases of extrahepatic cholangiocarcinoma.
RESULTS The expression levels of BAG-1 and BAD were measured with average absorbance (A) derived from the quantitative image analysis. Among 24, 13 and 5 cases of well, moderately and poorly differentiated extrahepatic cholangiocarcinomas, the average A of BAG-1 was respectively 0.068±0.037, 0.112±0.057 and 0.152±0.062, of BAD was respectively 0.079±0.053, 0.138±0.058 and 0.165±0.061, and the apoptotic indexes examined in the hematoxylin-and-eosin-stained specimens were respectively 1.1±0.5, 1.7±0.3 and 3.5±1.3. The expression levels of BAG-1 and BAD and the apoptotic indexes were increased obviously from well, moderately to poorly differentiated extrahepatic cholangiocarcinoma (P<0.05). The expression level of BAD was correlated to the apoptotic indexes (P<0.01).
CONCLUSION Expressions of BAG-1 and BAD and apoptosis indexes were all associated with differentiation of extrahepatic cholangiocarcinoma. BAG-1 and BAD may take part in the apoptosis modulation of the tumor.
Collapse
Affiliation(s)
- Qing-Guo Yan
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Jian-Guo Shi
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Gao-Sheng Huang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Chuan-Shan Zhang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Qing Li
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Pei-Zhen Hu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| | - Wen-Liang Wang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Province, China
| |
Collapse
|