1
|
Wang X, Zhang S, Han K, Wang L, Liu X. Induction of Apoptosis by Matrine Derivative ZS17 in Human Hepatocellular Carcinoma BEL-7402 and HepG2 Cells through ROS-JNK-P53 Signalling Pathway Activation. Int J Mol Sci 2022; 23:ijms232415991. [PMID: 36555631 PMCID: PMC9783520 DOI: 10.3390/ijms232415991] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies and ranks third among cancer-related deaths worldwide. Using matrine as a lead compound, 12 matrine derivatives were designed and synthesised, and their antiproliferative activities were evaluated in four cancer cell lines. Eight of the twelve compounds showed strong antiproliferative activity, with an IC50 of <10 μM. The compound ZS17 exhibited strong antiproliferative activity in hepatocellular carcinoma cell lines with IC50 values in the range of 3.014−3.388 μM, which was much lower than that of matrine. Furthermore, we explored the role of ZS17 in inducing apoptosis in HCC cells in vitro and in vivo, as well as possible mechanisms involved. ZS17 inhibited the proliferation of BEL-7402 and HepG2 cells in time- and dose-dependent manners. In addition, we found that ZS17 significantly induced apoptosis and ROS (reactive oxygen species) production, promoted JNK phosphorylation, activated p53, and activated the caspase signalling pathway. Furthermore, the antioxidant NAC, JNK inhibitor SP600125, and Si-JNK increased cell viability, re-established cell metastasis, and inhibited ZS17-induced apoptosis. An in vivo antitumour assay demonstrated that ZS17 significantly reduced the number of migrating HepG2 cells in zebrafish embryos and suppressed the growth of HepG2 xenografts in nude mice without any obvious side effects. Our study demonstrated that the ROS-JNK-P53 pathway plays an important role in the destruction of liver tumour cells by ZS17. Thus, ZS17 may represent a promising chemotherapeutic agent for the treatment of HCC patients.
Collapse
Affiliation(s)
| | | | | | | | - Xu Liu
- Correspondence: (L.W.); (X.L.)
| |
Collapse
|
2
|
Tsai CH, Lii CK, Wang TS, Liu KL, Chen HW, Huang CS, Li CC. Docosahexaenoic acid promotes the formation of autophagosomes in MCF-7 breast cancer cells through oxidative stress-induced growth inhibitor 1 mediated activation of AMPK/mTOR pathway. Food Chem Toxicol 2021; 154:112318. [PMID: 34116103 DOI: 10.1016/j.fct.2021.112318] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/13/2021] [Accepted: 05/29/2021] [Indexed: 11/18/2022]
Abstract
Docosahexaenoic acid (DHA) is known to regulate autophagy in cancer cells. We explored whether oxidative stress-induced growth inhibitor 1 (OSGIN1) is involved in the regulation of autophagy by DHA in breast cancer cells and the possible mechanisms involved. DHA upregulated the levels of OSGIN1, LC3-II and SQSTM1/p62. By contrast, DHA dose-dependently decreased the levels of mTOR and p-mTORS2448 expression. Using GFP/RFP-LC3 fluorescence staining, we showed that cells treated with DHA showed a dose-dependent response in autophagic signals. OSGIN1 Overexpression mimicked DHA treatment in that LC3-II and GFP/RFP-LC3 signals as well as the expression of p-AMPKαT172 and p-RaptorS792 were significantly increased, whereas mTOR, p-mTORS2448, and p-ULK1S757 expression were decreased. With knockdown of OSGIN1 expression, these outcomes were reversed. Moreover, OSGIN1 overexpression transiently elevated the accumulation of OSGIN1 and reactive oxygen species (ROS) in the mitochondrial fraction and subsequently increased p-AMPKαT172 and p-RaptorS792 expression. Upon pretreatment with Mito-TEMPO, a scavenger of mitochondrial ROS, these outcomes were reversed. Taken together, these results suggest that DHA can transiently elevate the generation of ROS in mitochondria and promote autophagosome formation through activation of the p-AMPKαT172/p-Raptor S792 and inactivation of the p-mTORS2448/p-ULK1Ser757 signaling pathways, and these effects depend on OSGIN1 protein in MCF-7 cells.
Collapse
Affiliation(s)
- Chia-Han Tsai
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Tsu-Shing Wang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chin-Shiu Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
3
|
Abubakar M, Fan S, Bowles EA, Widemann L, Duggan MA, Pfeiffer RM, Falk RT, Lawrence S, Richert-Boe K, Glass AG, Kimes TM, Figueroa JD, Rohan TE, Gierach GL. Relation of Quantitative Histologic and Radiologic Breast Tissue Composition Metrics With Invasive Breast Cancer Risk. JNCI Cancer Spectr 2021; 5:pkab015. [PMID: 33981950 PMCID: PMC8103888 DOI: 10.1093/jncics/pkab015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/09/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
Background Benign breast disease (BBD) is a strong breast cancer risk factor, but identifying patients that might develop invasive breast cancer remains a challenge. Methods By applying machine-learning to digitized hematoxylin and eosin-stained biopsies and computer-assisted thresholding to mammograms obtained circa BBD diagnosis, we generated quantitative tissue composition metrics and determined their association with future invasive breast cancer diagnosis. Archival breast biopsies and mammograms were obtained for women (18-86 years of age) in a case-control study, nested within a cohort of 15 395 BBD patients from Kaiser Permanente Northwest (1970-2012), followed through mid-2015. Patients who developed incident invasive breast cancer (ie, cases; n = 514) and those who did not (ie, controls; n = 514) were matched on BBD diagnosis age and plan membership duration. All statistical tests were 2-sided. Results Increasing epithelial area on the BBD biopsy was associated with increasing breast cancer risk (odds ratio [OR]Q4 vs Q1 = 1.85, 95% confidence interval [CI] = 1.13 to 3.04; P trend = .02). Conversely, increasing stroma was associated with decreased risk in nonproliferative, but not proliferative, BBD (P heterogeneity = .002). Increasing epithelium-to-stroma proportion (ORQ4 vs Q1 = 2.06, 95% CI =1.28 to 3.33; P trend = .002) and percent mammographic density (MBD) (ORQ4 vs Q1 = 2.20, 95% CI = 1.20 to 4.03; P trend = .01) were independently and strongly predictive of increased breast cancer risk. In combination, women with high epithelium-to-stroma proportion and high MBD had substantially higher risk than those with low epithelium-to-stroma proportion and low MBD (OR = 2.27, 95% CI = 1.27 to 4.06; P trend = .005), particularly among women with nonproliferative (P trend = .01) vs proliferative (P trend = .33) BBD. Conclusion Among BBD patients, increasing epithelium-to-stroma proportion on BBD biopsies and percent MBD at BBD diagnosis were independently and jointly associated with increasing breast cancer risk. These findings were particularly striking for women with nonproliferative disease (comprising approximately 70% of all BBD patients), for whom relevant predictive biomarkers are lacking.
Collapse
Affiliation(s)
- Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
- Correspondence to: Mustapha Abubakar, MD, PhD, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive, Rockville, MD, USA (e-mail: )
| | - Shaoqi Fan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Erin Aiello Bowles
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Lea Widemann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Máire A Duggan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Scott Lawrence
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | | | - Andrew G Glass
- Kaiser Permanente Center for Health Research, Portland, OR, USA
| | - Teresa M Kimes
- Kaiser Permanente Center for Health Research, Portland, OR, USA
| | - Jonine D Figueroa
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| |
Collapse
|
4
|
Yetkin Yildirim G, Tola EN, Dağ I. A novel biochemical marker - OKL38- with its apoptotic and antioxidant properties for the development of PCOS and its related clinical implications. Gynecol Endocrinol 2020; 36:673-677. [PMID: 31996062 DOI: 10.1080/09513590.2020.1718641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Aim: To investigate the role of serum OKL38 levels in the development of polycystic ovary syndrome (PCOS) and clinical implications related to PCOS.Method: PCOS (n = 33) and ovulatory controls (n = 48) were recruited for the study. Anthropometric measurements were recorded, and blood samples for hormonal and biochemical parameters including serum OKL38 levels were obtained. The potential role of OKL38 on the development of PCOS, metabolic syndrome and cardiovascular disease (CVD) were investigated. Framingham risk score (FRS) was used for the determination of CVD risk.Results: Mean Ferriman-Gallway (FG) score, insulin, low-density lipoprotein (LDL), total cholesterol (TC) levels, and the homeostasis model assessment of insulin resistance index (HOMA-IR) were significantly increased (p < .05) in women with PCOS compared to controls. PCOS group had lower mean OKL38 level compared to controls (p < .0001) and OKL38 was negatively predictive for the diagnosis of PCOS after adjustment of variables that were significantly different between two groups. A negative association between OKL38 and metabolic syndrome in PCOS women was evident after adjustment for age, obesity, and abdominal obesity. OKL38 level was also negatively correlated with body mass index, waist-to-hip-ratio, fat composition, serum TC, LDL, free testosterone levels, FRS, and FG scores.Conclusion: OKL38 may have a partial role in the etiopathogenesis of PCOS and may protect development of metabolic syndrome and CVD in women with PCOS.
Collapse
Affiliation(s)
- Gonca Yetkin Yildirim
- Department of Obstetrics and Gynecology, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Esra Nur Tola
- Department of Obstetrics and Gynecology, Istanbul Medipol University, Faculty of Medicine, Istanbul, Turkey
| | - Ismail Dağ
- Department of Biochemistry, Eyup State Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Zhang J, Wang L, Xu J, Tang Y, Huang B, Chen Z, Zhang T, Shen HM, Wu Y, Xia D. Bone marrow stromal cell-derived growth inhibitor serves as a stress sensor to induce autophagy. FEBS Lett 2020; 594:1248-1260. [PMID: 31945190 DOI: 10.1002/1873-3468.13732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/14/2019] [Accepted: 11/19/2019] [Indexed: 12/28/2022]
Abstract
Autophagy is an evolutionarily conserved stress response that promotes the lysosomal degradation of intracellular components. The bone marrow stromal cell-derived growth inhibitor (BDGI) functions as a stress sensor which is upregulated by oxidative stress and DNA damage. However, the role of BDGI in autophagic response to certain stresses remains unknown. Here, our results demonstrate that BDGI defines the impact of autophagy induction under stresses. Overexpression of BDGI promotes, while knockdown of BDGI impairs, autophagy. Mechanistically, BDGI localizes to the nucleus and interacts with the transcription factor transcription factor EB to increase the expression of multiple autophagy- and lysosome-related genes. In addition, BDGI regulates autophagy in a p53-dependent manner. Furthermore, BDGI-induced autophagy enables cell survival under stress conditions. Taken together, our study demonstrates that BDGI is a stress sensor that positively regulates autophagy.
Collapse
Affiliation(s)
- Jianbin Zhang
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People's Hospital,, People's Hospital of Hangzhou Medical College, China
| | - Liming Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jian Xu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Central Library, School of Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University, Hangzhou, China
| | - Yancheng Tang
- School of Chinese Medicine, Hong Kong Baptist University, China
| | - Bo Huang
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhifeng Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yihua Wu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Matrine Exerts Hepatotoxic Effects via the ROS-Dependent Mitochondrial Apoptosis Pathway and Inhibition of Nrf2-Mediated Antioxidant Response. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1045345. [PMID: 31737162 PMCID: PMC6815593 DOI: 10.1155/2019/1045345] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/14/2019] [Accepted: 08/31/2019] [Indexed: 02/08/2023]
Abstract
Matrine, an alkaloid isolated from Sophora flavescens, possesses a wide range of pharmacological properties. However, the use of matrine in clinical practice is limited due to its toxic effects. The present study investigated the roles of mitochondria and reactive oxygen species (ROS) in matrine-induced liver injury. Our results showed that treatment of HL-7702 cells with matrine led to significant and concentration- and time-dependent reductions in their viability, as well as significant and concentration-dependent increases in the number of apoptotic cells and supernatant lactate dehydrogenase (LDH) activity. The treatment led to significant increases in the population of cells in S phase and significant reduction of cell proportion in G0/G1 and G2/M phases. It also significantly and concentration-dependently increased the levels of ROS and malondialdehyde (MDA) but significantly and concentration-dependently reduced superoxide dismutase (SOD) activity, level of reduced glutathione (GSH), and mitochondrial membrane potential (MMP). Matrine treatment significantly and concentration-dependently upregulated the expressions of Bax, p53, p-p53, p21, cyclin E, Fas, cleaved caspase-3, caspase-8, and caspase-9 proteins and downregulated the expressions of Bcl-2, cyclin-dependent kinase 2 (CDK2), and cyclin A. It also significantly promoted the cleavage of poly(ADP-ribose)polymerase (PARP), upregulated Kelch-like ECH-associated protein 1 (Keap1) expression, and downregulated the expressions of cellular total and nuclear Nrf2. Matrine significantly inhibited the expressions of downstream oxidoreductases (Heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductases 1 (NQO-1)) and enhanced the formation of Keap1/Nrf2 protein complex. These results show that the hepatotoxic effect of matrine is exerted via inhibition of Nrf2 pathway, activation of ROS-mediated mitochondrial apoptosis pathway, and cell cycle arrest at S phase. Pretreatment with N-acetyl cysteine (NAC) partially reversed matrine-induced hepatotoxicity.
Collapse
|
7
|
Tsai CH, Shen YC, Chen HW, Liu KL, Chang JW, Chen PY, Lin CY, Yao HT, Li CC. Docosahexaenoic acid increases the expression of oxidative stress-induced growth inhibitor 1 through the PI3K/Akt/Nrf2 signaling pathway in breast cancer cells. Food Chem Toxicol 2017; 108:276-288. [DOI: 10.1016/j.fct.2017.08.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
|
8
|
Wang G, Zhou H, Strulovici-Barel Y, Al-Hijji M, Ou X, Salit J, Walters MS, Staudt MR, Kaner RJ, Crystal RG. Role of OSGIN1 in mediating smoking-induced autophagy in the human airway epithelium. Autophagy 2017; 13:1205-1220. [PMID: 28548877 DOI: 10.1080/15548627.2017.1301327] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Enhanced macroautophagy/autophagy is recognized as a component of the pathogenesis of smoking-induced airway disease. Based on the knowledge that enhanced autophagy is linked to oxidative stress and the DNA damage response, both of which are linked to smoking, we used microarray analysis of the airway epithelium to identify smoking upregulated genes known to respond to oxidative stress and the DNA damage response. This analysis identified OSGIN1 (oxidative stress induced growth inhibitor 1) as significantly upregulated by smoking, in both the large and small airway epithelium, an observation confirmed by an independent small airway microarray cohort, TaqMan PCR of large and small airway samples and RNA-Seq of small airway samples. High and low OSGIN1 expressors have different autophagy gene expression patterns in vivo. Genome-wide correlation of RNAseq analysis of airway basal/progenitor cells showed a direct correlation of OSGIN1 mRNA levels to multiple classic autophagy genes. In vitro cigarette smoke extract exposure of primary airway basal/progenitor cells was accompanied by a dose-dependent upregulation of OSGIN1 and autophagy induction. Lentivirus-mediated expression of OSGIN1 in human primary basal/progenitor cells induced puncta-like staining of MAP1LC3B and upregulation of MAP1LC3B mRNA and protein and SQSTM1 mRNA expression level in a dose and time-dependent manner. OSGIN1-induction of autophagosome, amphisome and autolysosome formation was confirmed by colocalization of MAP1LC3B with SQSTM1 or CD63 (endosome marker) and LAMP1 (lysosome marker). Both OSGIN1 overexpression and knockdown enhanced the smoking-evoked autophagic response. Together, these observations support the concept that smoking-induced upregulation of OSGIN1 is one link between smoking-induced stress and enhanced-autophagy in the human airway epithelium.
Collapse
Affiliation(s)
- Guoqing Wang
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Haixia Zhou
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA.,b Department of Respiratory Medicine , West China Hospital Sichuan University , Sichuan , China
| | - Yael Strulovici-Barel
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Mohammed Al-Hijji
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA.,c Weill Cornell Medical College-Qatar , Doha , Qatar
| | - Xuemei Ou
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA.,b Department of Respiratory Medicine , West China Hospital Sichuan University , Sichuan , China
| | - Jacqueline Salit
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Matthew S Walters
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Michelle R Staudt
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Robert J Kaner
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA.,d Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Ronald G Crystal
- a Department of Genetic Medicine , Weill Cornell Medical College , New York , NY , USA
| |
Collapse
|
9
|
Brennan MS, Matos MF, Richter KE, Li B, Scannevin RH. The NRF2 transcriptional target, OSGIN1, contributes to monomethyl fumarate-mediated cytoprotection in human astrocytes. Sci Rep 2017; 7:42054. [PMID: 28181536 PMCID: PMC5299414 DOI: 10.1038/srep42054] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/04/2017] [Indexed: 01/10/2023] Open
Abstract
Dimethyl fumarate (DMF) is indicated for the treatment of relapsing multiple sclerosis and may exert therapeutic effects via activation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2) pathway. Following oral DMF administration, central nervous system (CNS) tissue is predominantly exposed to monomethyl fumarate (MMF), the bioactive metabolite of DMF, which can stabilize NRF2 and induce antioxidant gene expression; however, the detailed NRF2-dependent mechanisms modulated by MMF that lead to cytoprotection are unknown. Our data identify a mechanism for MMF-mediated cytoprotection in human astrocytes that functions in an OSGIN1-dependent manner, specifically via upregulation of the OSGIN1-61 kDa isoform. NRF2-dependent OSGIN1 expression induced P53 nuclear translocation following MMF administration, leading to cell-cycle inhibition and cell protection against oxidative challenge. This study provides mechanistic insight into MMF-mediated cytoprotection via NRF2, OSGIN1, and P53 in human CNS-derived cells and contributes to our understanding of how DMF may act clinically to ameliorate pathological processes in neurodegenerative disease.
Collapse
Affiliation(s)
- Melanie S Brennan
- Neurology Research, Biogen Inc., Cambridge, MA, 02142, USA.,Boston University School of Medicine, Boston, MA 02118, USA
| | - Maria F Matos
- Neurology Research, Biogen Inc., Cambridge, MA, 02142, USA
| | - Karl E Richter
- Neurology Research, Biogen Inc., Cambridge, MA, 02142, USA
| | - Bing Li
- Neurology Research, Biogen Inc., Cambridge, MA, 02142, USA
| | | |
Collapse
|
10
|
Dai L, Cao Y, Chen Y, Kaleeba JAR, Zabaleta J, Qin Z. Genomic analysis of xCT-mediated regulatory network: Identification of novel targets against AIDS-associated lymphoma. Oncotarget 2016; 6:12710-22. [PMID: 25860939 PMCID: PMC4494968 DOI: 10.18632/oncotarget.3710] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/10/2015] [Indexed: 02/06/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of primary effusion lymphoma (PEL), a rapidly progressing malignancy mostly arising in HIV-infected patients. Even under conventional chemotherapy, PEL continues to portend nearly 100% mortality within several months, which urgently requires novel therapeutic strategies. We have previously demonstrated that targeting xCT, an amino acid transporter for cystine/glutamate exchange, induces significant PEL cell apoptosis through regulation of multiple host and viral factors. More importantly, one of xCT selective inhibitors, Sulfasalazine (SASP), effectively prevents PEL tumor progression in an immune-deficient xenograft model. In the current study, we use Illumina microarray to explore the profile of genes altered by SASP treatment within 3 KSHV+ PEL cell-lines, and discover that many genes involved in oxidative stress/antioxidant defense system, apoptosis/anti-apoptosis/cell death, and cellular response to unfolded proteins/topologically incorrect proteins are potentially regulated by xCT. We further validate 2 downstream candidates, OSGIN1 (oxidative stress-induced growth inhibitor 1) and XRCC5 (X-ray repair cross-complementing protein 5), and evaluate their functional relationship with PEL cell survival/proliferation and chemoresistance, respectively. Together, our data indicate that targeting these novel xCT-regulated downstream genes may represent a promising new therapeutic strategy against PEL and/or other AIDS-related lymphoma.
Collapse
Affiliation(s)
- Lu Dai
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias of The Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA, USA
| | - Yueyu Cao
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias of The Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yihan Chen
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias of The Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Johnan A R Kaleeba
- Department of Microbiology and Immunology, Uniformed Services University of The Health Sciences, Bethesda, MD, USA
| | - Jovanny Zabaleta
- Department of Pediatrics, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA, USA
| | - Zhiqiang Qin
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias of The Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Microbiology/Immunology/Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA, USA
| |
Collapse
|
11
|
Rodd AL, Ververis K, Sayakkarage D, Khan AW, Rafehi H, Ziemann M, Loveridge SJ, Lazarus R, Kerr C, Lockett T, El-Osta A, Karagiannis TC, Bennett LE. RNA sequencing supports distinct reactive oxygen species-mediated pathways of apoptosis by high and low size mass fractions of Bay leaf (Lauris nobilis) in HT-29 cells. Food Funct 2015; 6:2507-24. [PMID: 26114728 DOI: 10.1039/c5fo00467e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Anti-proliferative and pro-apoptotic effects of Bay leaf (Laurus nobilis) in mammalian cancer and HT-29 adenocarcinoma cells have been previously attributed to effects of polyphenolic and essential oil chemical species. Recently, we demonstrated differentiated growth-regulating effects of high (HFBL) versus low molecular mass (LFBL) aqueous fractions of bay leaf and now confirm by comparative effects on gene expression, that HFBL and LFBL suppress HT-29 growth by distinct mechanisms. Induction of intra-cellular lesions including DNA strand breakage by extra-cellular HFBL, invoked the hypothesis that iron-mediated reactive oxygen species with capacity to penetrate cell membrane, were responsible for HFBL-mediated effects, supported by equivalent effects of HFBL in combination with γ radiation. Activities of HFBL and LFBL were interpreted to reflect differentiated responses to iron-mediated reactive oxygen species (ROS), occurring either outside or inside cells. In the presence of LFBL, apoptotic death was relatively delayed compared with HFBL. ROS production by LFBL mediated p53-dependent apoptosis and recovery was suppressed by promoting G1/S phase arrest and failure of cellular tight junctions. In comparison, intra-cellular anti-oxidant protection exerted by LFBL was absent for extra-cellular HFBL (likely polysaccharide-rich), which potentiated more rapid apoptosis by producing DNA double strand breaks. Differentiated effects on expression of genes regulating ROS defense and chromatic condensation by LFBL versus HFBL, were observed. The results support ferrous iron in cell culture systems and potentially in vivo, can invoke different extra-cellular versus intra-cellular ROS-mediated chemistries, that may be regulated by exogenous, including dietary species.
Collapse
Affiliation(s)
- Annabelle L Rodd
- Epigenomic Medicine, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Transcriptomic analysis to elucidate the molecular mechanisms that underlie feed efficiency in meat-type chickens. Mol Genet Genomics 2015; 290:1673-82. [PMID: 25782841 DOI: 10.1007/s00438-015-1025-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 03/02/2015] [Indexed: 01/20/2023]
Abstract
Feed efficiency phenotypes defined by genotypes or gene markers are unknown. To date, there are only limited studies on global gene expression profiling on feed efficiency. The objective of this study was to identify genes and pathways associated with residual feed intake (RFI) through transcriptional profiling of duodenum at two different ages in a chicken population divergently selected for low (LRFI) or high (HRFI) RFI. The global gene expression differences in LRFI and HRFI were assessed by the Affymetrix GeneChip(®) Chicken Genome Array and RT-PCR using duodenal tissue on days 35 and 42. The Ingenuity Pathway Analysis program was used to identify canonical and gene network pathways associated with RFI. A global view of gene expression differences between LRFI and HRFI suggest that RFI can be explained by differences in cell division, growth, proliferation and apoptosis, protein synthesis, lipid metabolism, and molecular transport of cellular molecules. Chickens selected for improved RFI achieve efficiency by reducing feed intake with a nominal or no change in weight gain by either up-regulating CD36, PPARα, HMGCS2, GCG or down-regulating PCSK2, CALB1, SAT1, and SGK1 genes within the lipid metabolism, small molecule biochemistry, molecular transport, cell death, and protein synthesis molecular and cellular functions. Chickens selected for reduced RFI via reduced feed intake with no change in weight gain achieve feed efficiency for growth by the up-regulation of genes that reduce appetite with increased cellular oxidative stress, prolonged cell cycle, DNA damage, and apoptosis in addition to increased oxidation of dietary fat and efficient fatty acids transported from the intestines.
Collapse
|
13
|
Luna Vital DA, Loarca-Piña G, Dia VP, de Mejía EG. Peptides extracted from common bean (Phaseolus vulgaris L.) non-digestible fraction caused differential gene expression of HCT116 and RKO human colorectal cancer cells. Food Res Int 2014. [DOI: 10.1016/j.foodres.2014.02.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
14
|
BAE SEUNGHEE, JEONG HYEJUNG, CHA HWAJUN, KIM KARAM, CHOI YEONGMIN, AN INSOOK, KOH HYEAJUNG, LIM DAEJIN, LEE SUJAE, AN SUNGKWAN. The hypoxia-mimetic agent cobalt chloride induces cell cycle arrest and alters gene expression in U266 multiple myeloma cells. Int J Mol Med 2012; 30:1180-6. [DOI: 10.3892/ijmm.2012.1115] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 07/27/2012] [Indexed: 11/05/2022] Open
|
15
|
Hu J, Yao H, Gan F, Tokarski A, Wang Y. Interaction of OKL38 and p53 in regulating mitochondrial structure and function. PLoS One 2012; 7:e43362. [PMID: 22912861 PMCID: PMC3422280 DOI: 10.1371/journal.pone.0043362] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 07/20/2012] [Indexed: 11/25/2022] Open
Abstract
The tumor suppressor p53 is a well-known transcription factor controlling the expression of its target genes involved in cell cycle and apoptosis. In addition, p53 also plays a direct proapoptotic role in mitochondria by regulating cytochrome c release. Recently, we identified a novel downstream target of p53, OKL38, which relocalizes from nucleus to mitochondria upon forced expression to induce apoptosis. However, the mechanism underlying OKL38 targeting to mitochondria and apoptosis induction remains unclear. Here, we found that OKL38 interacts with p53 to regulate mitochondria function. After DNA damage, OKL38 colocalizes with p53 to mitochondria in U2OS cells. Further, p53 and OKL38 are targeted to mitochondria in synergy: forced expression of OKL38 leads to p53 localization to mitochondria while the expression of a mitochondria enriched p53 polymorphic variant, p53R72, leads to OKL38 enrichment in mitochondria. Biochemical analyses found that OKL38 and p53 interact in vivo and in vitro via multiple domains. In cell biological assays, multiple regions of OKL38 mediate its mitochondria localization and induce mitochondria morphology changes. OKL38 induces formation of megamitochondria and increases cellular levels of reactive oxygen species. Furthermore, OKL38 induces cytochrome c release upon incubation with mitochondria. Taken together, our studies suggest that OKL38 regulates mitochondria morphology and functions during apoptosis together with p53.
Collapse
Affiliation(s)
- Jing Hu
- Department of Biochemistry and Molecular Biology, Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Hongjie Yao
- Department of Biochemistry and Molecular Biology, Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Fei Gan
- Department of Biochemistry and Molecular Biology, Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Anthony Tokarski
- Department of Biochemistry and Molecular Biology, Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Yanming Wang
- Department of Biochemistry and Molecular Biology, Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Kampa M, Notas G, Pelekanou V, Troullinaki M, Andrianaki M, Azariadis K, Kampouri E, Lavrentaki K, Castanas E. Early membrane initiated transcriptional effects of estrogens in breast cancer cells: First pharmacological evidence for a novel membrane estrogen receptor element (ERx). Steroids 2012; 77:959-67. [PMID: 22406407 DOI: 10.1016/j.steroids.2012.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 01/20/2023]
Abstract
The complexity of estrogen actions mainly relies to the presence of different identified receptors (ERα, ERβ, their isoforms, and GPR30/GPER) and their discrete cellular distribution. Depending on the localization of the receptor that mediates estrogen effects, nuclear and extra-nuclear actions have been described. The latter can trigger a number of signaling events leading also to transcriptional modifications. In an attempt to clarify the nature of the receptor(s) involved in the membrane initiated effect of estrogens on gene expression, we performed a whole transcriptome analysis of breast cancer cell lines with different receptor profiles (T47D, MCF7, MDA-MB-231, SK-BR-3). A pharmacological approach was conducted with the use of estradiol (E(2)) or membrane-impermeable E(2)-BSA in the absence or presence of a specific ERα-β or GPR30/GPER antagonist. Our results clearly show that in addition to the ERα isoforms and/or GPR30/GPER that mainly mediate the transcriptional effect of E(2)-BSA, there is a specific transcriptional signature (found in T47D and MCF-7 cells) suggesting the presence of an unidentified membrane ER element (ERx). Analysis of its signature and phenotypic verification revealed that important cell function such as apoptosis, transcriptional regulation, and growth factor signaling are associated with ERx.
Collapse
Affiliation(s)
- Marilena Kampa
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kong BW, Lee JY, Bottje WG, Lassiter K, Lee J, Foster DN. Genome-wide differential gene expression in immortalized DF-1 chicken embryo fibroblast cell line. BMC Genomics 2011; 12:571. [PMID: 22111699 PMCID: PMC3258366 DOI: 10.1186/1471-2164-12-571] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 11/23/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND When compared to primary chicken embryo fibroblast (CEF) cells, the immortal DF-1 CEF line exhibits enhanced growth rates and susceptibility to oxidative stress. Although genes responsible for cell cycle regulation and antioxidant functions have been identified, the genome-wide transcription profile of immortal DF-1 CEF cells has not been previously reported. Global gene expression in primary CEF and DF-1 cells was performed using a 4X44K chicken oligo microarray. RESULTS A total of 3876 differentially expressed genes were identified with a 2 fold level cutoff that included 1706 up-regulated and 2170 down-regulated genes in DF-1 cells. Network and functional analyses using Ingenuity Pathways Analysis (IPA, Ingenuity® Systems, http://www.ingenuity.com) revealed that 902 of 3876 differentially expressed genes were classified into a number of functional groups including cellular growth and proliferation, cell cycle, cellular movement, cancer, genetic disorders, and cell death. Also, the top 5 gene networks with intermolecular connections were identified. Bioinformatic analyses suggested that DF-1 cells were characterized by enhanced molecular mechanisms for cell cycle progression and proliferation, suppressing cell death pathways, altered cellular morphogenesis, and accelerated capacity for molecule transport. Key molecules for these functions include E2F1, BRCA1, SRC, CASP3, and the peroxidases. CONCLUSIONS The global gene expression profiles provide insight into the cellular mechanisms that regulate the unique characteristics observed in immortal DF-1 CEF cells.
Collapse
Affiliation(s)
- Byung-Whi Kong
- Department of Poultry Science, Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, Arkansas 72701, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Galluzzi L, Morselli E, Kepp O, Vitale I, Pinti M, Kroemer G. Mitochondrial liaisons of p53. Antioxid Redox Signal 2011; 15:1691-714. [PMID: 20712408 DOI: 10.1089/ars.2010.3504] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Mitochondria play a central role in cell survival and cell death. While producing the bulk of intracellular ATP, mitochondrial respiration represents the most prominent source of harmful reactive oxygen species. Mitochondria participate in many anabolic pathways, including cholesterol and nucleotide biosynthesis, yet also control multiple biochemical cascades that contribute to the programmed demise of cells. The tumor suppressor protein p53 is best known for its ability to orchestrate a transcriptional response to stress that can have multiple outcomes, including cell cycle arrest and cell death. p53-mediated tumor suppression, however, also involves transcription-independent mechanisms. Cytoplasmic p53 can physically interact with members of the BCL-2 protein family, thereby promoting mitochondrial membrane permeabilization. Moreover, extranuclear p53 can suppress autophagy, a major prosurvival mechanism that is activated in response to multiple stress conditions. Thirty years have passed since its discovery, and p53 has been ascribed with an ever-increasing number of functions. For instance, p53 has turned out to influence the cell's redox status, by transactivating either anti- or pro-oxidant factors, and to regulate the metabolic switch between glycolysis and aerobic respiration. In this review, we will analyze the mechanisms by which p53 affects the balance between the vital and lethal functions of mitochondria.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- INSERM U848, Institut Gustave Roussy, Pavillon de Recherche 1, Villejuif (Paris), France
| | | | | | | | | | | |
Collapse
|
19
|
Analysis of genomic profile in mouse lymphoma L5178Y cells exposed to food colorant gardenia yellow. BIOCHIP JOURNAL 2010. [DOI: 10.1007/s13206-010-4405-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Giannoni P, Scaglione S, Daga A, Ilengo C, Cilli M, Quarto R. Short-time survival and engraftment of bone marrow stromal cells in an ectopic model of bone regeneration. Tissue Eng Part A 2010; 16:489-99. [PMID: 19712045 DOI: 10.1089/ten.tea.2009.0041] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In tissue-engineered applications bone marrow stromal cells (BMSCs) are combined with scaffolds to target bone regeneration; animal models have been devised and the cells' long-term engraftment has been widely studied. However, in regenerated bone, the cell number is severely reduced with respect to the initially seeded BMSCs. This reflects the natural low cellularity of bone but underlines the selectivity of the differentiation processes. In this respect, we evaluated the short-term survival of BMSCs, transduced with the luciferase gene, after implantation of cell-seeded scaffolds in a nude mouse model. Cell proliferation/survival was assessed by bioluminescence imaging: light production was decreased by 30% on the first day, reaching a 50% loss within 48 h. Less than 5% of the initial signal remained after 2 months in vivo. Apoptotic BMSCs were detected within the first 2 days of implantation. Interestingly, the initial frequency of clonogenic progenitors matched the percentage of in vivo surviving cells. Cytokines and inflammation may contribute to the apoptotic onset at the implant milieu. However, preculturing cells with tumor necrosis factor alpha enhanced survival, allowing detection of 8.1% of the seeded BMSCs 2 months after implantation. Thus culturing conditions may reduce the apoptotic overload of seeded osteoprogenitors, strengthening the constructs' osteogenic potential.
Collapse
Affiliation(s)
- Paolo Giannoni
- Stem Cell Laboratory, Advanced Biotechnology Center, Genova, Italy.
| | | | | | | | | | | |
Collapse
|
21
|
Yang B, Wu X, Mao Y, Bao W, Gao L, Zhou P, Xie R, Zhou L, Zhu J. Dual-targeted antitumor effects against brainstem glioma by intravenous delivery of tumor necrosis factor-related, apoptosis-inducing, ligand-engineered human mesenchymal stem cells. Neurosurgery 2009; 65:610-24; discussion 624. [PMID: 19687708 DOI: 10.1227/01.neu.0000350227.61132.a7] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE We sought to explore the dual-targeted antitumor effects of membrane-spanned, tumor necrosis factor-related, apoptosis-inducing ligand (TRAIL)-engineered human mesenchymal stem cells (hMSCs) against brainstem gliomas. METHODS The migration capacity of hMSCs toward gliomas was studied by the Transwell system in vitro and by intravenous injection of hMSCs in glioma-bearing mice in vivo. MSCs were engineered with native full-length human TRAIL (hTRAIL) by a recombinant adeno-associated virus (rAAV) vector (rAAV-hTRAIL). The targeted antiglioma effect was analyzed by coculture of hTRAIL-engineered MSCs with glioma in vitro and by systematic delivery of hTRAIL-engineered MSCs to established human brainstem glioma xenografts. RESULTS We demonstrated systematically that transplanted MSCs migrated to a brainstem glioma with a high specificity. MSCs penetrated the vessels surrounding the tumor, then streamed in a chain pattern toward the glioma, eventually surrounding the tumor. Membrane-spanned, TRAIL-engineered MSCs not only expressed full-length TRAIL in MSC surface, but secreted some soluble TRAIL in medium. After being infected with rAAV-hTRAIL, hMSCs showed no increase in apoptosis. After coculture of hTRAIL-engineered MSCs and U87MG cells, the apoptosis of U87MG cells significantly increased more than soluble TRAIL-treated U87MG cells. Systematic delivery of hTRAIL-engineered MSCs to established human brainstem glioma xenografts resulted in the potent induction of apoptosis in gliomas, but not in normal brain and prolonged survival to 38.0 +/- 10.46 days compared with phosphate-buffered saline (16.0 +/- 0.66 days), soluble TRAIL (19.0 +/- 1.65 days), and hMSC-LacZ (14.0 +/- 0.59 days) treated groups. CONCLUSION Systematically transplanted MSCs migrated to gliomas with a high specificity. Systematic delivery of MSC-hTRAIL can prolong the survival of brainstem glioma-bearing mice, presumably through a dual-targeted effect of membrane-spanned, TRAIL-engineered MSCs in the tumor microenvironment. MSCs may be an effective vehicle for the targeted delivery of therapeutic agents to brainstem gliomas.
Collapse
Affiliation(s)
- Bojie Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Omura S, Koike E, Kobayashi T. Microarray analysis of gene expression in rat alveolar epithelial cells exposed to fractionated organic extracts of diesel exhaust particles. Toxicology 2009; 262:65-72. [DOI: 10.1016/j.tox.2009.05.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 05/01/2009] [Accepted: 05/10/2009] [Indexed: 12/24/2022]
|
23
|
Shen H, Qin H, Guo J. Cooperation of metallothionein and zinc transporters for regulating zinc homeostasis in human intestinal Caco-2 cells. Nutr Res 2009; 28:406-13. [PMID: 19083439 DOI: 10.1016/j.nutres.2008.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 12/20/2007] [Accepted: 02/08/2008] [Indexed: 11/15/2022]
Abstract
This investigation examined the effects of zinc status on cell proliferation and the synergic roles of the metallothionein (MT) and zinc transporter (ZnT) in the human colon adenocarcinoma cell line Caco-2. Cells were treated with 0 to 300 micromol/L ZnSO(4) or 0 to 10 micromol/L N,N,N',N'-tetrakis(2-phridylmethyl) ethylenediamine (TPEN). Cell proliferation was determined by MTT assay and apoptotic cells detected by flow cytometry (Hoechst 33258 dye). mRNA expression of MT1; ZnT1; zrt, irt-like protein 1, 4 (ZIP1, 4); and divalent metal transporter (DMT1) were determined by the reverse transcription polymerase chain reaction or real-time polymerase chain reaction. The results showed that either high or low zinc could inhibit the cell proliferation. The number of apoptotic cells increased with incremental increases in the concentrations of ZnSO(4) and TPEN. The mRNA expression of ZnT1 and MT1 responded significantly after 6 and 12 hours with 200 micromol/L zinc treatment, respectively, and increased gradually with zinc levels from 0 to 200 micromol/L. Compared with the unchanged ZIP1 mRNA expression, ZIP4 was closely dependent on TPEN treatment duration and concentration. The DMT1 mRNA expression was upregulated time-dependently but not concentration-dependently in the late TPEN treatment duration. The results suggest that ZIP4 and DMT1 mRNA expressions are susceptible to low extracellular zinc concentration and upregulated to enhance zinc absorption, whereas the ZnT1 and MT1 act as the key regulators under high zinc conditions to enhance the intracellular zinc efflux to maintain zinc homeostasis. We propose that in response to variations in zinc concentration, the cooperated regulative roles of ZnT1, MT1, DMT1, and ZIP4 contribute to zinc homeostasis.
Collapse
Affiliation(s)
- Hui Shen
- Department of Military Hygiene, Second Military Medical University, Shanghai 200433, PR China.
| | | | | |
Collapse
|
24
|
Tian Z, An N, Zhou B, Xiao P, Kohane IS, Wu E. Cytotoxic diarylheptanoid induces cell cycle arrest and apoptosis via increasing ATF3 and stabilizing p53 in SH-SY5Y cells. Cancer Chemother Pharmacol 2008; 63:1131-9. [PMID: 18836721 DOI: 10.1007/s00280-008-0832-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Accepted: 09/02/2008] [Indexed: 01/15/2023]
Abstract
PURPOSE The aim of the study is to dissect the cytotoxic mechanisms of 1-(4-hydroxy-3-methoxyphenyl)-7-(3,4-dihydroxyphenyl)-4E-en-3-heptanone (compound 1) in SH-SY5Y cells and therefore to provide new insight into neuroblastoma chemotherapy. METHODS Nine diarylheptanoids were isolated from Alpinia officinarum by chromatography and their cytotoxicity was evaluated by an MTS assay. Flow cytometry, BrdU incorporation assay and fluorescence staining were employed to investigate cytostatic and apoptotic effects induced by the compound 1. In addition, Western blot, qPCR and siRNA techniques were used to elucidate the molecular mechanisms of the cytotoxicity. RESULTS The study to elucidate the cytotoxic mechanisms of compound 1, the most potent diarylheptanoid showed that cell cycle-related proteins, cyclins, CDKs and CDKIs, as well as two main apoptotic related families, caspase and Bcl 2 were involved in S phase arrest and apoptosis in neuroblastoma cell line SH-SY5Y. Furthermore, following the drug treatment, the protein expression of p53, phospho-p53 (Ser20) as well as the p53 transcriptional activated genes ATF3, puma and Apaf-1 were increased dramatically; MDM2 and Aurora A, the two p53 negative regulators were decreased; the p53 protein stability was enhanced, whereas the p53 mRNA expression level slightly decreased and ATF3 mRNA expression apparently increased. In addition, the knockdown of ATF3 gene by siRNA partially suppressed p53, caspase 3, S phase arrest and apoptosis triggered by compound 1. CONCLUSION These results suggest that compound 1 induces S phase arrest and apoptosis via up regulation of ATF3 and stabilization of p53 in SH-SY5Y cell line. Therefore, compound 1 might be a promising lead structure for neuroblastoma therapy.
Collapse
Affiliation(s)
- Ze Tian
- Informatics Program, MIT Division of Health Sciences and Technology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
25
|
Yao H, Li P, Venters BJ, Zheng S, Thompson PR, Pugh BF, Wang Y. Histone Arg modifications and p53 regulate the expression of OKL38, a mediator of apoptosis. J Biol Chem 2008; 283:20060-8. [PMID: 18499678 PMCID: PMC2459274 DOI: 10.1074/jbc.m802940200] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 05/21/2008] [Indexed: 11/06/2022] Open
Abstract
Protein Arg methyltransferases function as coactivators of the tumor suppressor p53 to regulate gene expression. Peptidylarginine deiminase 4 (PAD4/PADI4) counteracts the functions of protein Arg methyltransferases in gene regulation by deimination and demethylimination. Here we show that the expression of a tumor suppressor gene, OKL38, is activated by the inhibition of PAD4 or the activation of p53 following DNA damage. Chromatin immunoprecipitation assays showed a dynamic change of p53 and PAD4 occupancy and histone Arg modifications at the OKL38 promoter during DNA damage, suggesting a direct role of PAD4 and p53 in the expression of OKL38. Furthermore, we found that OKL38 induces apoptosis through localization to mitochondria and induction of cytochrome c release. Together, our studies identify OKL38 as a novel p53 target gene that is regulated by PAD4 and plays a role in apoptosis.
Collapse
Affiliation(s)
- Hongjie Yao
- Center for Gene Regulation, Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Wang X, Liu B, Li N, Li H, Qiu J, Zhang Y, Cao X. IPP5, a novel protein inhibitor of protein phosphatase 1, promotes G1/S progression in a Thr-40-dependent manner. J Biol Chem 2008; 283:12076-84. [PMID: 18310074 DOI: 10.1074/jbc.m801571200] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein phosphatase 1 (PP1) is a major serine/threonine phosphatase that controls gene expression and cell cycle progression. Here we describe the characterization of a novel inhibitory molecule for PP1, human inhibitor-5 of protein phosphatase 1 (IPP5). We find that IPP5, containing the PP1 inhibitory subunits, specifically interacts with the PP1 catalytic subunit and inhibits PP1 phosphatase activity. Furthermore, the mutation of Thr-40 within the inhibitory subunit of IPP5 into Ala eliminates the phosphorylation of IPP5 by protein kinase A and its inhibitor activity to PP1, whereas the mutation of Thr-40 within a truncated form of IPP5 into Asp can serve as a dominant active form of IPP5 in inhibiting PP1 activity. In IPP5-negative SW480 and IPP5-highly positive SW620 human colon cancer cells, we find that overexpression of IPP5 promotes the growth and accelerates the G(1)-S transition of SW480 cells in a Thr-40-dependent manner, which could be reversed by downregulation of the PP1 expression. Moreover, silencing of IPP5 inhibits the growth of SW620 cells both in vitro and in nude mice possibly by inducing G(0)/G(1) arrest but not by promoting apoptosis. According to its role in the promotion of cell cycle progression and cell growth, IPP5 up-regulates the expression of cyclin E and the phosphorylated form of retinoblastoma protein. Our findings suggest that IPP5, by acting as an inhibitory molecule for PP1, can promote tumor cell growth and cell cycle progression, and may be a promising target in cancer therapeutics in IPP5-highly expressing tumor cells.
Collapse
Affiliation(s)
- Xiaojian Wang
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Rui Y, Liu X, Li N, Jiang Y, Chen G, Cao X, Wang J. PECAM-1 ligation negatively regulates TLR4 signaling in macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:7344-51. [PMID: 18025177 DOI: 10.4049/jimmunol.179.11.7344] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Uncontrolled TLR4 signaling may induce excessive production of proinflammatory cytokines and lead to harmful inflammation; therefore, negative regulation of TLR4 signaling attracts much attention now. PECAM-1, a member of Ig-ITIM family, can mediate inhibitory signals in T cells and B cells. However, the role and the mechanisms of PECAM-1 in the regulation of TLR4-mediated LPS response in macrophages remain unclear. In this study, we demonstrate that PECAM-1 ligation with CD38-Fc fusion protein negatively regulates LPS-induced proinflammatory cytokine TNF-alpha, IL-6, and IFN-beta production by inhibiting JNK, NF-kappaB, and IFN regulatory factor 3 activation in macrophages. In addition, PECAM-1 ligation-recruited Src homology region 2 domain-containing phosphatase 1 (SHP-1) and Src homology region 2 domain-containing phosphatase 2 (SHP-2) may be involved in the inhibitory effect of PECAM-1 on TLR4 signaling. Consistently, silencing of PECAM-1 enhances the macrophage response to LPS stimulation. Taken together with the data that PECAM-1 is constitutively expressed in macrophages and its expression is up-regulated by LPS stimulation, PECAM-1 might function as a feedback negative regulator of LPS inflammatory response in macrophages. This study may provide a potential target for intervention of inflammatory diseases.
Collapse
Affiliation(s)
- Yuxiang Rui
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Tian Z, Shen J, Moseman AP, Yang Q, Yang J, Xiao P, Wu E, Kohane IS. Dulxanthone A induces cell cycle arrest and apoptosisvia up-regulation of p53 through mitochondrial pathway in HepG2 cells. Int J Cancer 2007; 122:31-8. [PMID: 17847033 DOI: 10.1002/ijc.23048] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Natural products derived from plants provide a rich source for development of new anticancer drugs. Dulxanthone A was found to be an active cytotoxic component in Garcinia cowa by bioactivity-directed isolation. Studies to elucidate the cytotoxic mechanisms of dulxanthone A showed that dulxanthone A consistently induced S phase arrest and apoptosis in the most sensitive cell line HepG2. Furthermore, p53 was dramatically up-regulated, leading to altered expression of downstream proteins upon dulxanthone A treatment. Cell cycle related proteins, such as cyclin A, cyclin B, cyclin E, cdc-2, p21 and p27 were down-regulated. Some apoptosis correlated proteins were also altered following the drug treatment. Bcl-2 family members PUMA was up-regulated while Bcl-2 and Bax were down-regulated. However, the expression ratio of Bax/Bcl-2 was increased. This resulted in the release of cytochrome C from the mitochondria to the cytosol. Concurrently, Apaf-1 was stimulated with p53 by dulxanthone A. In result, cytochrome C, Apaf-1 and procaspase-9 form an apoptosome, which in turn triggered the activation of caspase-9, caspase-3 and downstream caspase substrates. Lamin A/C and PARP were down-regulated or cleaved, respectively. Moreover, cell cycle arrest and apoptosis in HepG2 cells induced by dulxanthone A were markedly inhibited by siRNA knockdown of p53. In summary, dulxanthone A is an active cytotoxic component of G. cowa. It induces cell cycle arrest at lower concentrations and triggers apoptosis at higher concentrations via up-regulation of p53 through the intrinsic mitochondrial pathway in HepG2 cells. Dulxanthone A is therefore likely a promising preventive and/or therapeutic agent against Hepatoma.
Collapse
Affiliation(s)
- Ze Tian
- Children's Hospital Informatics Program at Harvard- MIT Division of Health Sciences and Technology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Ong CK, Leong C, Tan PH, Van T, Huynh H. The role of 5′ untranslated region in translational suppression of OKL38 mRNA in hepatocellular carcinoma. Oncogene 2006; 26:1155-65. [PMID: 16924236 DOI: 10.1038/sj.onc.1209896] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide. OKL38 is a pregnancy-induced growth inhibitory gene and its expression is lost in various breast cancer cell lines and kidney tumor. To determine the role of OKL38 expression in HCC, we investigated its expression in various HCC samples and liver cancer cell lines. Western blot analysis revealed that OKL38 protein was absent or reduced in 64.2% (18 of 28) of the HCCs examined and four liver cancer cell lines. Immunohistochemistry study demonstrated that OKL38 protein was undetectable in 41.3% (38 of 92) of HCC, whereas 39.1% (36 of 92) of HCC showed low expression of the protein. Lost or reduced expression level of OKL38 protein was significantly correlated to high tumor stages in HCC (P=0.0042). Overexpression of the OKL38 caused cell death in Chang liver cells. 5' Untranslated region (5'UTR) deletion studies demonstrated that OKL38 was downregulated via translation suppression associated with the 5'UTR of its mRNA. Taken together, the 5'UTRs of OKL38 might play an important role in downregulation of its protein and the absence of OKL38 could lead to the development or progression of HCC.
Collapse
Affiliation(s)
- C K Ong
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, National Cancer Centre of Singapore, Singapore, Singapore
| | | | | | | | | |
Collapse
|
30
|
Li HC, Stoicov C, Rogers AB, Houghton J. Stem cells and cancer: Evidence for bone marrow stem cells in epithelial cancers. World J Gastroenterol 2006; 12:363-71. [PMID: 16489634 PMCID: PMC4066053 DOI: 10.3748/wjg.v12.i3.363] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer commonly arises at the sites of chronic inflammation and infection. Although this association has long been recognized, the reason has remained unclear. Within the gastrointestinal tract, there are many examples of inflammatory conditions associated with cancer, and these include reflux disease and Barrett’s adenocarcinoma of the esophagus, Helicobacter infection and gastric cancer, inflammatory bowel disease and colorectal cancer and viral hepatitis leading to hepatocellular carcinoma. There are several mechanisms by which chronic inflammation has been postulated to lead to cancer which includes enhanced proliferation in an endless attempt to heal damage, the presence of a persistent inflammatory environment creating a pro-carcinogenic environment and more recently a role for engraftment of circulating marrow-derived stem cells which may contribute to the stromal components of the tumor as well as the tumor mass itself. Here we review the recent advances in our understanding of the contributions of circulating bone marrow-derived stem cells to the formation of tumors in animal models as well as in human beings.
Collapse
Affiliation(s)
- Han-Chen Li
- Department of Medicine, University of Massachusetts Medical School, LRB-Second Floor, Room 209, 364 Plantation Street, Worcester, MA 01605-2324, United States
| | | | | | | |
Collapse
|
31
|
Abstract
Cells derived from bone marrow are pluripotent, with the ability to differentiate into multiple cell types. Environmental cues dictate differentiation decisions. It should not be surprising then, that abnormal cell environments lead to abnormal differentiation of these cells, and in some cases, malignant transformation. Identifying a role for bone marrow-derived cells in the initiation and progression of cancer allows a dramatic change in the way in which cancer is viewed. Identifying the cell responsible for initiating a tumor offers the exciting possibility of specifically targeting unique aspects of these cells and altering signaling properties for more effective therapeutic approaches.
Collapse
Affiliation(s)
- Calin Stoicov
- University of Massachusetts Medical School, Department of Medicine, Division of Gasteroenterology, Department of Cancer Biology, Worcester MA 01605, USA
| | | | | | | |
Collapse
|
32
|
Chen W, Li N, Chen T, Han Y, Li C, Wang Y, He W, Zhang L, Wan T, Cao X. The lysosome-associated apoptosis-inducing protein containing the pleckstrin homology (PH) and FYVE domains (LAPF), representative of a novel family of PH and FYVE domain-containing proteins, induces caspase-independent apoptosis via the lysosomal-mitochondrial pathway. J Biol Chem 2005; 280:40985-95. [PMID: 16188880 DOI: 10.1074/jbc.m502190200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lysosomes have recently been identified as important apoptotic signal integrators in response to various stimuli. Here we report the functional characterization of LAPF, a novel lysosome-associated apoptosis-inducing protein containing PH and FYVE domains. LAPF is a representative of a new protein family, the Phafins (protein containing both PH and FYVE domains), which consists of 14 unidentified proteins from various species. Overexpression of LAPF in L929 cells induces apoptosis and also increases cell sensitivity to TNFalpha-induced apoptosis, concomitant with its translocation to lysosomes. Two mutants of LAPF, either lacking the PH or FYVE domain, failed to induce cell death and translocate to lysosomes, suggesting that both domains are required for its apoptosis-inducing activity and relocation. We demonstrate that LAPF may induce apoptosis via the following steps: LAPF translocation to lysosomes, lysosomal membrane permeabilization (LMP), release of cathepsin (cath) D and L, mitochondrial membrane permeabilization (MMP), release of apoptosis-inducing factor (AIF), and caspase-independent apoptosis. The cath D-specific inhibitor attenuates LAPF-induced apoptosis, indicating a pivotal role of lysosomes in LAPF-initiated apoptosis. We also demonstrate that the lysosomal pathway was employed in the typical apoptotic model in which high dose TNFalpha was used to stimulate L929 cells. Silencing of LAPF expression by small RNA interference protected L929 cells from hTNFalpha-induced apoptosis by impairing hTNFalpha-triggered LMP and MMP. Therefore, LAPF may launch caspase-independent apoptosis through the lysosomal-mitochondrial pathway.
Collapse
Affiliation(s)
- Wei Chen
- Institute of Immunology, Zhejiang University, 353 Yanan Road, Hangzhou 310031, Zhejiang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|