1
|
Guo M, Li X, Tao W, Teng F, Li C. Vibrio splendidus infection promotes circRNA-FGL1-regulated coelomocyte apoptosis via competitive binding to Myc with the deubiquitinase OTUB1 in Apostichopus japonicus. PLoS Pathog 2024; 20:e1012463. [PMID: 39146353 PMCID: PMC11349225 DOI: 10.1371/journal.ppat.1012463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/27/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Abstract
Circular RNAs (circRNAs) are involved in various physiological and pathological processes in both vertebrates and invertebrates. However, most studies on circRNAs have focused on their roles as endogenous competitive RNAs. Here, we report a novel function of circRNA derived from the Fibrinogen-like protein 1 gene (circ-FGL1) that inhibits coelomocyte apoptosis via competing with the deubiquitinase AjOTUB1 to bind AjMyc in Apostichopus japonicus during Vibrio splendidus infection. The results showed that circ-FGL1 is significantly downregulated in coelomocytes of V. splendidus-induced A. japonicus and negatively regulates coelomocyte apoptosis through the AjBax-AjCyt c pathway. Mechanistically, the deubiquitinase AjOTUB1 and circ-FGL1 could interact with the transcription factor protein AjMyc in the same region with circ-FGL1/AjMyc having greater affinity. Under normal conditions, high levels of circ-FGL1 bind directly to AjMyc, inhibiting the deubiquitylation of AjMyc by AjOTUB1 and leading to the degradation of AjMyc. After V. splendidus infection, AjMyc disassociates from the depressed expression of circ-FGL1, promoting its deubiquitylation by binding to the induced deubiquitinase AjOTUB1 to inhibit its degradation. AjMyc is then transferred to the nucleus and promotes the transcription of AjCyt c and AjBax to induce coelomocyte apoptosis. The new finding will expand our present outstanding on the functional role of circRNAs and suggest new therapeutic targets for the treatment of echinoderms during bacterial invasion.
Collapse
Affiliation(s)
- Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Xin Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Wenjun Tao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Fei Teng
- College of Mathematics and Computer, Jilin Normal University, Siping, Jilin, China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
2
|
Diepstraten ST, Young S, La Marca JE, Wang Z, Kluck RM, Strasser A, Kelly GL. Lymphoma cells lacking pro-apoptotic BAX are highly resistant to BH3-mimetics targeting pro-survival MCL-1 but retain sensitivity to conventional DNA-damaging drugs. Cell Death Differ 2023; 30:1005-1017. [PMID: 36755070 PMCID: PMC10070326 DOI: 10.1038/s41418-023-01117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
BH3-mimetic drugs are an anti-cancer therapy that can induce apoptosis in malignant cells by directly binding and inhibiting pro-survival proteins of the BCL-2 family. The BH3-mimetic drug venetoclax, which targets BCL-2, has been approved for the treatment of chronic lymphocytic leukaemia and acute myeloid leukaemia by regulatory authorities worldwide. However, while most patients initially respond well, resistance and relapse while on this drug is an emerging and critical issue in the clinic. Though some studies have begun uncovering the factors involved in resistance to BCL-2-targeting BH3-mimetic drugs, little focus has been applied to pre-emptively tackle resistance for the next generation of BH3-mimetic drugs targeting MCL-1, which are now in clinical trials for diverse blood cancers. Therefore, using pre-clinical mouse and human models of aggressive lymphoma, we sought to predict factors likely to contribute to the development of resistance in patients receiving MCL-1-targeting BH3-mimetic drugs. First, we performed multiple whole genome CRISPR/Cas9 KO screens and identified that loss of the pro-apoptotic effector protein BAX, but not its close relative BAK, could confer resistance to MCL-1-targeting BH3-mimetic drugs in both short-term and long-term treatment regimens, even in lymphoma cells lacking the tumour suppressor TRP53. Furthermore, we found that mouse Eµ-Myc lymphoma cells selected for loss of BAX, as well as upregulation of the untargeted pro-survival BCL-2 family proteins BCL-XL and A1, when made naturally resistant to MCL-1 inhibitors by culturing them in increasing doses of drug over time, a situation mimicking the clinical application of these drugs. Finally, we identified therapeutic approaches which could overcome these two methods of resistance: the use of chemotherapeutic drugs or combined BH3-mimetic treatment, respectively. Collectively, these results uncover some key factors likely to cause resistance to MCL-1 inhibition in the clinic and suggest rational therapeutic strategies to overcome resistance that should be investigated further.
Collapse
Affiliation(s)
- Sarah T Diepstraten
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Savannah Young
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - John E La Marca
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Zilu Wang
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Ruth M Kluck
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
3
|
Stiller KT, Kolarevic J, Lazado CC, Gerwins J, Good C, Summerfelt ST, Mota VC, Espmark ÅMO. The Effects of Ozone on Atlantic Salmon Post-Smolt in Brackish Water-Establishing Welfare Indicators and Thresholds. Int J Mol Sci 2020; 21:E5109. [PMID: 32698319 PMCID: PMC7404298 DOI: 10.3390/ijms21145109] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/03/2020] [Accepted: 07/12/2020] [Indexed: 12/20/2022] Open
Abstract
Ozone is a strong oxidant, and its use in aquaculture has been shown to improve water quality and fish health. At present, it is predominantly used in freshwater systems due to the high risk of toxic residual oxidant exposure in brackish water and seawater. Here, we report the effects of ozone on Atlantic salmon (Salmo salar) post-smolts (~100 g), in a brackish water (12 ppt) flow-through system. Salmon were exposed to oxidation reduction potential concentrations of 250 mV (control), 280 mV (low), 350 mV (medium), 425 mV (high) and 500 mV (very high). The physiological impacts of ozone were characterized by blood biochemical profiling, histopathologic examination and gene expression analysis in skin and gills. Fish exposed to 425 mV and higher showed ≥33% cumulative mortality in less than 10 days. No significant mortalities were recorded in the remaining groups. The skin surface quality and the thickness of the dermal and epidermal layers were not significantly affected by the treatments. On the other hand, gill histopathology showed the adverse effects of increasing ozone doses and the changes were more pronounced in the group exposed to 350 mV and higher. Cases of gill damages such as necrosis, lamellar fusion and hypertrophy were prevalent in the high and very high groups. Expression profiling of key biomarkers for mucosal health supported the histology results, showing that gills were significantly more affected by higher ozone doses compared to the skin. Increasing ozone doses triggered anti-oxidative stress and inflammatory responses in the gills, where transcript levels of glutathione reductase, copper/zinc superoxide dismutase, interleukin 1β and interleukin were significantly elevated. Heat shock protein 70 was significantly upregulated in the skin of fish exposed to 350 mV and higher. Bcl-2 associated x protein was the only gene marker that was significantly upregulated by increasing ozone doses in both mucosal tissues. In conclusion, the study revealed that short-term exposure to ozone at concentrations higher than 350 mV in salmon in brackish water resulted in significant health and welfare consequences, including mortality and gill damages. The results of the study will be valuable in developing water treatment protocols for salmon farming.
Collapse
Affiliation(s)
- Kevin T. Stiller
- Nofima AS, NO 9291 Tromsø, Norway; (J.K.); (C.C.L.); (J.G.); (V.C.M.); (Å.M.O.E.)
| | - Jelena Kolarevic
- Nofima AS, NO 9291 Tromsø, Norway; (J.K.); (C.C.L.); (J.G.); (V.C.M.); (Å.M.O.E.)
| | - Carlo C. Lazado
- Nofima AS, NO 9291 Tromsø, Norway; (J.K.); (C.C.L.); (J.G.); (V.C.M.); (Å.M.O.E.)
| | - Jascha Gerwins
- Nofima AS, NO 9291 Tromsø, Norway; (J.K.); (C.C.L.); (J.G.); (V.C.M.); (Å.M.O.E.)
| | - Christopher Good
- The Conservation Fund’s Freshwater Institute, Shepherdstown, WV 25443, USA; (C.G.); (S.T.S.)
| | - Steven T. Summerfelt
- The Conservation Fund’s Freshwater Institute, Shepherdstown, WV 25443, USA; (C.G.); (S.T.S.)
| | - Vasco C. Mota
- Nofima AS, NO 9291 Tromsø, Norway; (J.K.); (C.C.L.); (J.G.); (V.C.M.); (Å.M.O.E.)
| | - Åsa M. O. Espmark
- Nofima AS, NO 9291 Tromsø, Norway; (J.K.); (C.C.L.); (J.G.); (V.C.M.); (Å.M.O.E.)
| |
Collapse
|
4
|
Zhang Y, Shao Y, Lv Z, Li C. MYC regulates coelomocytes apoptosis by targeting Bax expression in sea cucumber Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2020; 97:27-33. [PMID: 31843700 DOI: 10.1016/j.fsi.2019.12.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/28/2019] [Accepted: 12/12/2019] [Indexed: 06/10/2023]
Abstract
Myelocytomatosis viral oncogene (MYC), a multifunctional transcription factor, (TF) exerts various physiological and pathological effects on animals. AjMYC could induce coelomocyte apoptosis in Apostichopus japonicus, but the underlying molecular mechanism remains poorly understood. In this study, the promoter sequence of apoptosis regulator Bcl-2-associated X (Bax) was cloned by genomic walking. The AjBax promoter region spaning 1189 bp, containing several transcription factor binding sites, included four potential E-boxes (-1030 bp to -1019 bp, -785 bp to -774 bp, -570 bp to -559 bp, -100 bp to -89 bp), two P53 binding sites (-439 bp to -430 bp, -845 bp to -836 bp), and one NF-κB site (-191 bp to -182 bp). Transient transfection of EPC cells with 5'-deletion constructs linked to luciferase reporter revealed that the region -1189/+454 contributed importantly to the expression of the AjBax. In addition, the AjBax promoter was induced by LPS, PGN or MAN. The four potential MYC binding sites were cotransfected with AjMYC in EPC cell whether AjMYC could activate AjBax expression as a transcriptional factor. Only P1 (-1189/+454) fragment containing the first MYC binding site transfection increased the luciferase activity by 2.08-fold (p < 0.01) compared with the control. The first MYC binding site -1030/-1019 was essential to induce AjBax transcription. Further functional assay indicated that AjBax was significantly induced by 3.54-fold increase (p < 0.01) after AjMYC overexpression in sea cucumber coelomocytes. All our findings supported that AjMYC could regulate coelomocyte apoptosis by directly targeting AjBax expression in A. japonicus.
Collapse
Affiliation(s)
- Yi Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Zhimeng Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
5
|
Martin LJ, Chang Q. DNA Damage Response and Repair, DNA Methylation, and Cell Death in Human Neurons and Experimental Animal Neurons Are Different. J Neuropathol Exp Neurol 2018; 77:636-655. [PMID: 29788379 PMCID: PMC6005106 DOI: 10.1093/jnen/nly040] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurological disorders affecting individuals in infancy to old age elude interventions for meaningful protection against neurodegeneration, and preclinical work has not translated to humans. We studied human neuron responses to injury and death stimuli compared to those of animal neurons in culture under similar settings of insult (excitotoxicity, oxidative stress, and DNA damage). Human neurons were differentiated from a cortical neuron cell line and the embryonic stem cell-derived H9 line. Mouse neurons were differentiated from forebrain neural stem cells and embryonic cerebral cortex; pig neurons were derived from forebrain neural stem cells. Mitochondrial morphology was different in human and mouse neurons. Human and mouse neurons challenged with DNA-damaging agent camptothecin showed different chromatin condensation, cell death, and DNA damage sensor activation. DNA damage accumulation and repair kinetics differed among human, mouse, and pig neurons. Promoter CpG island methylation microarrays showed significant differential DNA methylation in human and mouse neurons after injury. Therefore, DNA damage response, DNA repair, DNA methylation, and autonomous cell death mechanisms in human neurons and experimental animal neurons are different.
Collapse
Affiliation(s)
- Lee J Martin
- Department of Pathology, Division of Neuropathology
- Pathobiology Graduate Training Program
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qing Chang
- Department of Pathology, Division of Neuropathology
| |
Collapse
|
6
|
Sarma P, Bag I, Ramaiah MJ, Kamal A, Bhadra U, Pal Bhadra M. Bisindole-PBD regulates breast cancer cell proliferation via SIRT-p53 axis. Cancer Biol Ther 2015; 16:1486-501. [PMID: 26192233 DOI: 10.1080/15384047.2015.1071731] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In a previous study we reported the role of potent bisindole-PBD conjugate as an inclusion in the arsenal of breast cancer therapeutics. In breast cancer cell proliferation, PI3K/AKT/mTOR pathway plays a crucial role by prosurvival mechanism that inhibits programmed cell death. Here, 2 breast cancer cells lines, MCF-7 and MDA-MB-231 were treated with Vorinostat (suberoylanilide hydroxamic acid / SAHA) and bisindole-PBD (5b). We have investigated the effect on PI3K/AKT/mTOR pathway and SIRT expression including epigenetic regulation. There was consistent decrease in the level of PI3K, AKT, mTOR proteins upon treatment of 5b in both MCF-7 and MDA-MB-231 cell lines compared to untreated controls. Treatment with caspase inhibitor (Q-VD-OPH) confirmed that the effect of 5b on PI3K signaling was ahead of apoptosis. Real time PCR and western blot analysis showed profound reduction in the mRNA and protein levels of SIRT1 and SIRT2. Molecular docking studies also supported the interaction of 5b with various amino acids of SIRT2 proteins. Treatment with 5b caused epigenetic changes that include increase of acetylated forms of p53, increase of histone acetylation at p21 promoter as well as decrease in methylation state of p21 gene. Compound 5b thus acts as SIRT inhibitor and cause p53 activation via inhibition of growth factor signaling and activation of p53 dependent apoptotic signaling. This present study focuses bisindole-PBD on epigenetic alteration putting 5b as a promising therapeutic tool in the realm of breast cancer research.
Collapse
Affiliation(s)
- Pranjal Sarma
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India
| | - Indira Bag
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India.,b Functional Genomics and Gene Silencing Group; CSIR-Center for Cellular and Molecular Biology ; Hyderabad , India
| | - M Janaki Ramaiah
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India.,c School of Chemical & Biotechnology; SASTRA University ; Tirumalaisamudram, Thanjavur , India
| | - Ahmed Kamal
- d Medicinal Chemistry and Pharmacology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India
| | - Utpal Bhadra
- b Functional Genomics and Gene Silencing Group; CSIR-Center for Cellular and Molecular Biology ; Hyderabad , India
| | - Manika Pal Bhadra
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India
| |
Collapse
|
7
|
Bodur C, Kutuk O, Karsli-Uzunbas G, Isimjan TT, Harrison P, Basaga H. Pramanicin analog induces apoptosis in human colon cancer cells: critical roles for Bcl-2, Bim, and p38 MAPK signaling. PLoS One 2013; 8:e56369. [PMID: 23441183 PMCID: PMC3575438 DOI: 10.1371/journal.pone.0056369] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/08/2013] [Indexed: 11/19/2022] Open
Abstract
Pramanicin (PMC) is an antifungal agent that was previously demonstrated to exhibit antiangiogenic and anticancer properties in a few in vitro studies. We initially screened a number of PMC analogs for their cytotoxic effects on HCT116 human colon cancer cells. PMC-A, the analog with the most potent antiproliferative effect was chosen to further interrogate the underlying mechanism of action. PMC-A led to apoptosis through activation of caspase-9 and -3. The apoptotic nature of cell death was confirmed by abrogation of cell death with pretreatment with specific caspase inhibitors. Stress-related MAPKs JNK and p38 were both activated concomittantly with the intrinsic apoptotic pathway. Moreover, pharmacological inhibition of p38 proved to attenuate the cell death induction while pretreatment with JNK inhibitor did not exhibit a protective effect. Resistance of Bax −/− cells and the protective nature of caspase-9 inhibition indicate that mitochondria play a central role in PMC-A induced apoptosis. Early post-exposure elevation of cellular Bim and Bax was followed by a marginal Bcl-2 depletion and Bid cleavage. Further analysis revealed that Bcl-2 downregulation occurs at the mRNA level and is critical to mediate PMC-A induced apoptosis, as ectopic Bcl-2 expression substantially spared the cells from death. Conversely, forced expression of Bim proved to significantly increase cell death. In addition, analyses of p53−/− cells demonstrated that Bcl-2/Bim/Bax modulation and MAPK activations take place independently of p53 expression. Taken together, p53-independent transcriptional Bcl-2 downregulation and p38 signaling appear to be the key modulatory events in PMC-A induced apoptosis.
Collapse
Affiliation(s)
- Cagri Bodur
- Biological Sciences and Bioengineering Program, Sabanci University, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
8
|
Yu HJ, Shin JA, Nam JS, Kang BS, Cho SD. Apoptotic effect of dibenzylideneacetone on oral cancer cells via modulation of specificity protein 1 and Bax. Oral Dis 2013; 19:767-74. [DOI: 10.1111/odi.12062] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 12/11/2012] [Accepted: 12/19/2012] [Indexed: 12/11/2022]
Affiliation(s)
- H-J Yu
- Department of Oral Pathology; School of Dentistry; Institute of Oral Bioscience; Chonbuk National University; Jeonju; Korea
| | - J-A Shin
- Department of Oral Pathology; School of Dentistry; Institute of Oral Bioscience; Chonbuk National University; Jeonju; Korea
| | - J-S Nam
- Laboratory of Tumor suppressor; Lee Gil Ya Cancer and Diabetes Institute; Gachon University; Inchon; Korea
| | - B S Kang
- Bio-medical Research Institute; Kyungpook National University Hospital; Daegu; Korea
| | - S-D Cho
- Department of Oral Pathology; School of Dentistry; Institute of Oral Bioscience; Chonbuk National University; Jeonju; Korea
| |
Collapse
|
9
|
Lu HP, Li J, Mo WJ, Feng ZB. RNAi-mediated down-regulation of Sp3 gene expression inhibits proliferation of HepG2 cells. Shijie Huaren Xiaohua Zazhi 2012; 20:2595-2600. [DOI: 10.11569/wcjd.v20.i27.2595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [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 effect of RNA interference (RNAi)-mediated gene silencing of specificity protein 3 (Sp3) on the proliferation of human hepatocellular carcinoma HepG2 cells.
METHODS: HepG2 cells were infected with a lentivirus expressing Sp3-siRNA, and the expression of Sp3 mRNA and protein was determined by semi-quantitative reverse transcription polymerase chain reaction (RT-PCR), and Western blot and immunohistochemistry. Cell growth was evaluated by methyl thiazolyl tetrazolium (MTT) assay, and cell cycle progression was analyzed by flow cytometry.
RESULTS: Compared to control cells, the expression levels of Sp3 mRNA and protein were significantly lower in HepG2 cells transfected with the Sp3-siRNA (mRNA: 0.47 ± 0.05 vs 0.74 ± 0.08, 0.70 ± 0.16, F = 7.322, all P < 0.05; protein: 0.37 ± 0.08vs 0.83 ± 0.17, 0.66 ± 0.13, F = 8.442, all P < 0.05). MTT assay showed that the growth of cells transfected with the Sp3-siRNA was slower at 48 , 72 and 96 h (0.28 ± 0.18 vs 0.34 ± 0.19, 0.35 ± 0.07, F = 3.888; 0.57 ± 0.11 vs 0.84 ± 0.05, 0.74 ± 0.08, F = 12.721; 0.72 ± 18.1 vs 0.98 ± 0.05, 0.93 ± 0.9, F = 6.342, all P < 0.05). Flow cytometry analysis showed that the percentage of cells in G1 phase increased in cells transfected with the Sp3-siRNA.
CONCLUSION: Sp3 may play an important role in the growth of human hepatic cancer cells, and RNAi-induced Sp3 down-regulation could inhibit the growth of HepG2 cells in vitro.
Collapse
|
10
|
Gu H, Li D, Sung CK, Yim H, Troke P, Benjamin T. DNA-binding and regulatory properties of the transcription factor and putative tumor suppressor p150(Sal2). BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1809:276-83. [PMID: 21362508 DOI: 10.1016/j.bbagrm.2011.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 02/10/2011] [Accepted: 02/22/2011] [Indexed: 12/26/2022]
Abstract
The product of the SALL2 protein p150(Sal2) is a multi-zinc finger transcription factor with growth arrest and proapoptotic functions that overlap those of p53. Its DNA-binding properties are unknown. We have used a modified SELEX procedure with purified p150(Sal2) and a pool of oligonucleotides of random sequence to identify those that are bound preferentially by p150(Sal2). The consensus sequence for optimal binding in vitro is GGG(T/C)GGG, placing p150(Sal2) among a large group of GC box-binding proteins including the Sp1 family of transcription factors. A triple zinc finger motif in p150(Sal2) similar to that in Sp1 is required for DNA binding. p150(Sal2) and Sp1 show evidence of co-operative binding in vitro and of interaction in vivo. p150(Sal2), a known activator of the CDK inhibitor p21(Cip1/Waf1) (p21), binds to regions of the human p21 promoter that contain variations of the consensus sequence in multiple copies. p150(Sal2) is also shown to bind to the BAX promoter with similar elements and to activate its expression following an apoptotic stimulus. These results demonstrate binding of p150(Sal2) to two natural promoters with GC elements related to the optimal binding sequence defined in vitro and whose regulation is important for suppression of tumor growth.
Collapse
Affiliation(s)
- Hongcang Gu
- Department of pathology NRB-939, Harvard Medical School, 77 Avenue Pasteur, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
11
|
A single nucleotide polymorphism in the Bax gene promoter affects transcription and influences retinal ganglion cell death. ASN Neuro 2010; 2:e00032. [PMID: 20360947 PMCID: PMC2847828 DOI: 10.1042/an20100003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 02/24/2010] [Accepted: 03/02/2010] [Indexed: 11/17/2022] Open
Abstract
Pro-apoptotic Bax is essential for RGC (retinal ganglion cell)
death. Gene dosage experiments in mice, yielding a single wild-type
Bax allele, indicated that genetic background was able to
influence the cell death phenotype. DBA/2JBax+/− mice exhibited complete resistance to nerve damage after 2 weeks
(similar to Bax−/− mice), but 129B6Bax+/− mice exhibited significant cell loss (similar to wild-type mice). The
different cell death phenotype was associated with the level of
Bax expression, where 129B6 neurons had twice the level of
endogenous Bax mRNA and protein as DBA/2J neurons. Sequence
analysis of the Bax promoters between these strains revealed a
single nucleotide polymorphism (T129B6 to CDBA/2J) at
position −515. A 1.5- to 2.5-fold increase in transcriptional
activity was observed from the 129B6 promoter in transient transfection assays
in a variety of cell types, including RGC5 cells derived from rat RGCs. Since
this polymorphism occurred in a p53 half-site, we investigated the requirement
of p53 for the differential transcriptional activity. Differential
transcriptional activity from either 129B6 or DBA/2J Bax
promoters were unaffected in p53−/− cells, and
addition of exogenous p53 had no further effect on this difference, thus a role
for p53 was excluded. Competitive electrophoretic mobility-shift assays
identified two DNA–protein complexes that interacted with the
polymorphic region. Those forming Complex 1 bound with higher affinity to the
129B6 polymorphic site, suggesting that these proteins probably comprised a
transcriptional activator complex. These studies implicated quantitative
expression of the Bax gene as playing a possible role in
neuronal susceptibility to damaging stimuli.
Collapse
|
12
|
Martin LJ, Liu Z, Pipino J, Chestnut B, Landek MA. Molecular regulation of DNA damage-induced apoptosis in neurons of cerebral cortex. Cereb Cortex 2008; 19:1273-93. [PMID: 18820287 DOI: 10.1093/cercor/bhn167] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cerebral cortical neuron degeneration occurs in brain disorders manifesting throughout life, but the mechanisms are understood poorly. We used cultured embryonic mouse cortical neurons and an in vivo mouse model to study mechanisms of DNA damaged-induced apoptosis in immature and differentiated neurons. p53 drives apoptosis of immature and differentiated cortical neurons through its rapid and prominent activation stimulated by DNA strand breaks induced by topoisomerase-I and -II inhibition. Blocking p53-DNA transactivation with alpha-pifithrin protects immature neurons; blocking p53-mitochondrial functions with mu-pifithrin protects differentiated neurons. Mitochondrial death proteins are upregulated in apoptotic immature and differentiated neurons and have nonredundant proapoptotic functions; Bak is more dominant than Bax in differentiated neurons. p53 phosphorylation is mediated by ataxia telangiectasia mutated (ATM) kinase. ATM inactivation is antiapoptotic, particularly in differentiated neurons, whereas inhibition of c-Abl protects immature neurons but not differentiated neurons. Cell death protein expression patterns in mouse forebrain are mostly similar to cultured neurons. DNA damage induces prominent p53 activation and apoptosis in cerebral cortex in vivo. Thus, DNA strand breaks in cortical neurons induce rapid p53-mediated apoptosis through actions of upstream ATM and c-Abl kinases and downstream mitochondrial death proteins. This molecular network operates through variations depending on neuron maturity.
Collapse
Affiliation(s)
- Lee J Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2196, USA.
| | | | | | | | | |
Collapse
|
13
|
Kanamori H, Takemura G, Li Y, Okada H, Maruyama R, Aoyama T, Miyata S, Esaki M, Ogino A, Nakagawa M, Ushikoshi H, Kawasaki M, Minatoguchi S, Fujiwara H. Inhibition of Fas-associated apoptosis in granulation tissue cells accompanies attenuation of postinfarction left ventricular remodeling by olmesartan. Am J Physiol Heart Circ Physiol 2007; 292:H2184-94. [PMID: 17208988 DOI: 10.1152/ajpheart.01235.2006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Blockade of angiotensin II type 1 receptor (AT1) signaling attenuates heart failure following myocardial infarction (MI), perhaps through reduction of fibrosis in the noninfarcted myocardium. However, its specific effect on the infarct tissue itself has not been fully clarified, which we examined in the present study. After MI induction in mice, treatment with the AT1 blocker olmesartan, beginning on the 3rd day post-MI, significantly improved survival (94%) 4 wk post-MI, compared with saline (53%) and hydralazine (73%). Olmesartan-treated mice also showed significant attenuation of left ventricular dilatation and dysfunction, as well as significantly greater infarct wall thickness, although the absolute size of the infarct scar was unchanged. In addition, significantly greater numbers of nonmyocytes (mainly vascular cells and myofibroblasts) were present within the infarct scar in olmesartan-treated hearts. Ten days post-MI, apoptosis among granulation tissue cells was significantly suppressed in the olmesartan-treated hearts, where expression of Fas, Bax, procaspase-3, and Daxx and activation of caspase-3, c-Jun NH2-terminal kinase, and c-Jun were all significantly attenuated. By contrast, expression of Fas ligand, Bcl-2, and Fas-associated death domain and activation of caspase-8 were unaffected, suggesting olmesartan exerts a negative regulatory effect on the alternate pathway downstream of Fas receptor. In vitro, olmesartan dose-dependently inhibited Fas-mediated apoptosis in granulation tissue-derived myofibroblasts. The present study proposes this antiapoptotic effect as another important mechanism for an AT1 blocker in improving post-MI ventricular remodeling, as well as its antifibrotic effect, and also suggests a significant link between renin-angiotensin and Fas/Fas ligand systems in postinfarction hearts.
Collapse
Affiliation(s)
- Hiromitsu Kanamori
- Second Department of Internal Medicine, Gifu University School of Medicine, 1-1 Yanagito, Gifu 501-1194, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
di Masi A, Antoccia A, Dimauro I, Argentino-Storino A, Mosiello A, Mango R, Novelli G, Tanzarella C. Gene expression and apoptosis induction in p53-heterozygous irradiated mice. Mutat Res 2006; 594:49-62. [PMID: 16169021 DOI: 10.1016/j.mrfmmm.2005.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 07/11/2005] [Accepted: 07/29/2005] [Indexed: 05/04/2023]
Abstract
The role of the p53-genetic background in the expression of genes involved in either cell cycle checkpoint activation or apoptosis was evaluated in p53+/+ and p53+/- mouse strains at both basal levels and after DNA-induced damage. The spleen, colon, kidneys, lungs and liver of both strains were harvested from untreated animals and from mice exposed to 7.5 Gy of X-rays and sacrificed after 5 h. No significant differences were observed in the basal levels of p53 protein, CDKN1A and bax mRNA and spontaneous apoptosis, neither among the different organs within the same strain, nor between the same organ in the p53+/+ and p53+/- strains. After X-ray exposure, p53-dependent regulation was strikingly tissue-specific. In wild-type irradiated mice, p53 protein level increased after radiation treatment in all the organs analysed, whereas both CDKN1A and bax genes transcription increased in the spleen, colon and lungs, as assessed by means of quantitative RT-PCR. In p53+/- irradiated mice, on the contrary, a significant p53 induction was detected only in the spleen, while CDKN1A and bax genes levels increased in the spleen, colon and lungs, revealing the existence of different mechanisms of gene regulation in different organs. Apoptosis induction was observed in the spleen and colon of both strains, even if to lower extent in p53+/- mice compared to p53+/+ animals. In conclusion, in the spleen and colon, target gene transcription and apoptosis may be related to p53 genotype after DNA damage-induction. Moreover, our findings highlight the selectivity of p53 in transactivation following DNA damage in vivo, resulting in tissue-specific responses.
Collapse
Affiliation(s)
- Alessandra di Masi
- Department of Biology, University of Rome Roma Tre, Viale G. Marconi, 446, 00146 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Lou Z, O'Reilly S, Liang H, Maher VM, Sleight SD, McCormick JJ. Down-Regulation of Overexpressed Sp1 Protein in Human Fibrosarcoma Cell Lines Inhibits Tumor Formation. Cancer Res 2005. [DOI: 10.1158/0008-5472.1007.65.3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Sp1 is a transcription factor for many genes, including genes involved in tumorigenesis. We found that human fibroblast cells malignantly transformed in culture by a carcinogen or by stable transfection of an oncogene express Sp1 at 8-fold to 18-fold higher levels than their parental cells. These cell lines form fibrosarcomas in athymic mice with a very short latency, and the cells from the tumors express the same high levels of Sp1. Similar high levels of Sp1 were found in the patient-derived fibrosarcoma cell lines tested, and in the tumors formed in athymic mice by these cell lines. To investigate the role of overexpression of Sp1 in malignant transformation of human fibroblasts, we transfected an Sp1 U1snRNA/Ribozyme into two human cell lines, malignantly transformed in culture by a carcinogen or overexpression of an oncogene, and into a patient-derived fibrosarcoma cell line. The level of expression of Sp1 in these transfected cell lines was reduced to near normal. The cells regained the spindle-shaped morphology and exhibited increased apoptosis and decreased expression of several genes linked to cancer, i.e., epithelial growth factor receptor, urokinase plasminogen activator, urokinase plasminogen activator receptor, and vascular endothelial growth factor. When injected into athymic mice, these cell lines with near normal levels of Sp1 failed to form tumors or did so only at a greatly reduced frequency and with a much longer latency. These data indicate that overexpression of Sp1 plays a causal role in malignant transformation of human fibroblasts and suggest that for cancers in which it is overexpressed, Sp1 constitutes a target for therapy.
Collapse
Affiliation(s)
- Zhenjun Lou
- Carcinogenesis Laboratory, Department of Microbiology and Molecular Genetics and Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Sandra O'Reilly
- Carcinogenesis Laboratory, Department of Microbiology and Molecular Genetics and Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Hongyan Liang
- Carcinogenesis Laboratory, Department of Microbiology and Molecular Genetics and Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Veronica M. Maher
- Carcinogenesis Laboratory, Department of Microbiology and Molecular Genetics and Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Stuart D. Sleight
- Carcinogenesis Laboratory, Department of Microbiology and Molecular Genetics and Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - J. Justin McCormick
- Carcinogenesis Laboratory, Department of Microbiology and Molecular Genetics and Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
16
|
Firlej V, Bocquet B, Desbiens X, de Launoit Y, Chotteau-Lelièvre A. Pea3 Transcription Factor Cooperates with USF-1 in Regulation of the Murine bax Transcription without Binding to an Ets-binding Site. J Biol Chem 2005; 280:887-98. [PMID: 15466854 DOI: 10.1074/jbc.m408017200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Pea3 transcription factor (which belongs to the PEA3 group) from the Ets family has been shown to be involved in mammary embryogenesis and oncogenesis. However, except for proteinases, only few of its target genes have been reported. In the present report, we identified bax as a Pea3 up-regulated gene. We provide evidence of this regulation by using Pea3 overexpression and Pea3 silencing in a mammary cell line. Both Pea3 and Erm, another member of the PEA3 group, are able to transactivate bax promoter fragments. Although the minimal Pea3-regulated bax promoter does not contain an Ets-binding site, two functional upstream stimulatory factor-regulated E boxes are present. We further demonstrate the ability of Pea3 and USF-1 to cooperate for the transactivation of the bax promoter, mutation of the E boxes dramatically reducing the Pea3 transactivation potential. Although Pea3 did not directly bind to the minimal bax promoter, we provide evidence that USF-1 could form a ternary complex with Pea3 and DNA. Taken together, our results suggest that Pea3 may regulate bax transcription via the interaction with USF-1 but without binding to DNA.
Collapse
Affiliation(s)
- Virginie Firlej
- Laboratoire de Biologie du Développement UPRES-EA1033, Université des Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq, France
| | | | | | | | | |
Collapse
|
17
|
Yakovlev AG, Di Giovanni S, Wang G, Liu W, Stoica B, Faden AI. BOK and NOXA Are Essential Mediators of p53-dependent Apoptosis. J Biol Chem 2004; 279:28367-74. [PMID: 15102863 DOI: 10.1074/jbc.m313526200] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cellular stress leads to DNA damage and activation of the intrinsic apoptotic pathway in which translocation of mitochondrial cytochrome c to the cytosol plays a critical role. Previous studies have suggested alternative mechanisms responsible for this process. We examined initiation mechanisms of the intrinsic apoptotic pathway using human neuroblastoma and breast cancer cells. Results indicated that translocation of cytochrome c does not require prior activation of caspases but rather depends on activation of specific BCL-2 family members, depending upon the type of death signal. Thus, DNA damage-induced apoptosis requires new protein synthesis, accumulation of p53 tumor suppressor protein, and p53-dependent induction of BOK and NOXA genes, while a role for BAX in this pathway is not essential. In contrast, apoptosis induced by staurosporine does not require protein synthesis but is characterized by translocation of BAX. Based on these findings, we propose a model of the intrinsic apoptotic cascade induced by DNA damage where proapoptotic BOK substitutes for a function of BAX.
Collapse
Affiliation(s)
- Alexander G Yakovlev
- Department of Neuroscience, Georgetown University, Research Building WP-14, 3970 Reservoir Road NW, Washington, D. C. 20007, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Kaufmann SH, Vaux DL. Alterations in the apoptotic machinery and their potential role in anticancer drug resistance. Oncogene 2003; 22:7414-30. [PMID: 14576849 DOI: 10.1038/sj.onc.1206945] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Anticancer drugs can potentially kill cells in two fundamentally different ways, by interfering with cellular processes that are essential for maintenance of viability or by triggering an endogenous physiological cell death mechanism. Apoptosis is a form of physiological cell death mediated by caspases, a unique family of intracellular cysteine proteases. Zymogen forms of these proteases are found in virtually all somatic cells, but remain latent until their activation is induced by ligation of specific cell surface receptors (the so-called "death receptors"), by mitochondrial alterations that allow release of cytochrome c and other intermembrane components, or possibly by other mechanisms. Most anticancer drugs activate the mitochondrial pathway. This apoptotic pathway is regulated by pro- and antiapoptotic members of the Bcl-2 family of proteins. Once activated, certain caspases might also be controlled by the inhibitor of apoptosis (IAP) proteins. Alterations in apoptotic pathway components or their regulators have been detected in a variety of cancers, suggesting that loss of the ability of cells to undergo apoptosis might contribute to carcinogenesis. Because cancer therapies such as radiation, glucocorticoids, and chemotherapeutic drugs exert their beneficial effects, at least in part, by inducing apoptosis of cancer cells, the same alterations in apoptotic pathways would be predicted to contribute to resistance. A key issue is whether the direct toxic activity of these treatments is of benefit when neoplastic cells contain changes that diminish their ability to undergo apoptosis.
Collapse
Affiliation(s)
- Scott H Kaufmann
- Division of Oncology Research, Guggenheim 1342C, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | |
Collapse
|
19
|
Kohl A, Clayton RF, Weber F, Bridgen A, Randall RE, Elliott RM. Bunyamwera virus nonstructural protein NSs counteracts interferon regulatory factor 3-mediated induction of early cell death. J Virol 2003; 77:7999-8008. [PMID: 12829839 PMCID: PMC161919 DOI: 10.1128/jvi.77.14.7999-8008.2003] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2003] [Accepted: 04/21/2003] [Indexed: 02/03/2023] Open
Abstract
The genome of Bunyamwera virus (BUN; family Bunyaviridae, genus Orthobunyavirus) consists of three segments of negative-sense RNA. The smallest segment, S, encodes two proteins, the nonstructural protein NSs, which is nonessential for viral replication and transcription, and the nucleocapsid protein N. Although a precise role in the replication cycle has yet to be attributed to NSs, it has been shown that NSs inhibits the induction of alpha/beta interferon, suggesting that it plays a part in counteracting the host antiviral defense. A defense mechanism to limit viral spread is programmed cell death by apoptosis. Here we show that a recombinant BUN that does not express NSs (BUNdelNSs) induces apoptotic cell death more rapidly than wild-type virus. Screening for apoptosis pathways revealed that the proapoptotic transcription factor interferon regulatory factor 3 (IRF-3) was activated by both wild-type BUN and BUNdelNSs infection, but only wild-type BUN was able to suppress signaling downstream of IRF-3. Studies with a BUN minireplicon system showed that active replication induced an IRF-3-dependent promoter, which was suppressed by the NSs protein. In a cell line (P2.1) defective in double-stranded RNA signaling due to low levels of IRF-3, induction of apoptosis was similar for wild-type BUN and BUNdelNSs. These data suggest that the BUN NSs protein can delay cell death in the early stages of BUN infection by inhibiting IRF-3-mediated apoptosis.
Collapse
Affiliation(s)
- Alain Kohl
- Division of Virology, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G11 5JR, Scotland, United Kingdom
| | | | | | | | | | | |
Collapse
|
20
|
Kim E, Deppert W. The complex interactions of p53 with target DNA: we learn as we go. Biochem Cell Biol 2003; 81:141-50. [PMID: 12897847 DOI: 10.1139/o03-046] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The most import biological function of the tumor suppressor p53 is that of a sequence-specific transactivator. In response to a variety of cellular stress stimuli, p53 induces the transcription of an ever-increasing number of target genes, leading to growth arrest and repair, or to apoptosis. Long considered as a "latent" DNA binder that requires prior activation by C-terminal modification, recent data provide strong evidence that the DNA binding activity of p53 is strongly dependent on structural features within the target DNA and is latent only if the target DNA lacks a certain structural signal code. In this review we discuss evidence for complex interactions of p53 with DNA, which are strongly dependent on the dynamics of DNA structure, especially in the context of chromatin. We provide a model of how this complexity may serve to achieve selectivity of target gene regulation by p53 and how DNA structure in the context of chromatin may serve to modulate p53 functions.
Collapse
Affiliation(s)
- Ella Kim
- Heinrich-Pette-Institut für Experimentelle Virologie und Immunologie, Universität Hamburg, Germany
| | | |
Collapse
|
21
|
Thornborrow EC, Patel S, Mastropietro AE, Schwartzfarb EM, Manfredi JJ. A conserved intronic response element mediates direct p53-dependent transcriptional activation of both the human and murine bax genes. Oncogene 2002; 21:990-9. [PMID: 11850816 DOI: 10.1038/sj.onc.1205069] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2001] [Revised: 10/05/2001] [Accepted: 10/12/2001] [Indexed: 11/09/2022]
Abstract
Both the human and the mouse bax promoters contain p53 binding sites which are sufficient to confer p53-dependent transcriptional activation in a heterologous setting. Nevertheless in the context of the bax promoter, these sites do not mediate a p53-dependent response, suggesting that bax may not be a direct transcriptional target of p53. Here, data are presented identifying a conserved p53 response element in the first intron of both the human and the murine bax genes. This element both in isolation and in the context of the first intron conferred p53-dependent transcriptional activation upon a minimal promoter. Electrophoretic mobility shift assays demonstrated that this sequence also is capable of mediating sequence specific binding to p53. p53 effectively activated transcription through both human and murine bax gene reporter constructs, whereas deletion of the intronic response element abrogated the p53-responsiveness of both reporters. Interestingly, tumor-derived mutants of p53 which are defective in inducing an apoptotic response retain the ability to activate transcription via the bax intronic p53 site. Since these mutants are transcriptionally inactive on the p53 site in the bax promoter, the ability of these mutants to up-regulating endogenous bax mRNA levels supports a role for the intronic element in p53-dependent up-regulation of bax expression. Taken together, these results show the requirement for a novel intronic element in the p53-dependent transcriptional activation of bax, and demonstrate that bax is indeed a direct and evolutionarily conserved transcriptional target of p53.
Collapse
Affiliation(s)
- Edward C Thornborrow
- Derald H Ruttenberg Cancer Center, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
22
|
Lawrence T, Gilroy DW, Colville-Nash PR, Willoughby DA. Possible new role for NF-kappaB in the resolution of inflammation. Nat Med 2001; 7:1291-7. [PMID: 11726968 DOI: 10.1038/nm1201-1291] [Citation(s) in RCA: 592] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Inflammation involves the sequential activation of signaling pathways leading to the production of both pro- and anti-inflammatory mediators. Although much attention has focused on pro-inflammatory pathways that initiate inflammation, relatively little is known about the mechanisms that switch off inflammation and resolve the inflammatory response. The transcription factor NF-kappaB is thought to have a central role in the induction of pro-inflammatory gene expression and has attracted interest as a new target for the treatment of inflammatory disease. We show here that NF-kappaB activation in leukocytes recruited during the onset of inflammation is associated with pro-inflammatory gene expression, whereas such activation during the resolution of inflammation is associated with the expression of anti-inflammatory genes and the induction of apoptosis. Inhibition of NF-kappaB during the resolution of inflammation protracts the inflammatory response and prevents apoptosis. This suggests that NF-kappaB has an anti-inflammatory role in vivo involving the regulation of inflammatory resolution.
Collapse
Affiliation(s)
- T Lawrence
- Department of Experimental Pathology, William Harvey Research Institute, Charterhouse Square, London EC1M 6BQ, UK.
| | | | | | | |
Collapse
|
23
|
Chen JY, Hsu PC, Hsu IL, Yeh GC. Sequential up-regulation of the c-fos, c-jun and bax genes in the cortex, striatum and cerebellum induced by a single injection of a low dose of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in C57BL/6 mice. Neurosci Lett 2001; 314:49-52. [PMID: 11698144 DOI: 10.1016/s0304-3940(01)02281-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated whether single injection of 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) (20 mg/kg) will alter the expression of pro-apoptotic genes, namely, the c-fos, c-jun, and bax, in the striatum, cortex, and cerebellum of adult male C57BL/6 mice using reverse transcription-polymerase chain reaction assay. Injection of MPTP induced a transient decrease in the content of tyrosine hydroxylase estimated by the immunoreactivity in the striatum, which completely recovered 14 day after injection. A rapid but transient up-regulation of c-fos and c-jun genes occurred an hour after MPTP-injection, and a delayed but persistent up-regulation of bax gene expression occurred 3 day after injection. The up-regulation of these genes was present in all the examined brain regions. This result suggests that MPTP, at a low dose causing transient degeneration in the striatum, is capable of triggering two genetic pathways related to the generation of apoptosis in both dopaminergic and non-dopaminergic systems in the mouse brain.
Collapse
Affiliation(s)
- J Y Chen
- Department of Pediatrics, Taipei Medical University, Taipei 110, Taiwan
| | | | | | | |
Collapse
|
24
|
Matikainen T, Perez GI, Jurisicova A, Pru JK, Schlezinger JJ, Ryu HY, Laine J, Sakai T, Korsmeyer SJ, Casper RF, Sherr DH, Tilly JL. Aromatic hydrocarbon receptor-driven Bax gene expression is required for premature ovarian failure caused by biohazardous environmental chemicals. Nat Genet 2001; 28:355-60. [PMID: 11455387 DOI: 10.1038/ng575] [Citation(s) in RCA: 316] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are toxic chemicals released into the environment by fossil fuel combustion. Moreover, a primary route of human exposure to PAHs is tobacco smoke. Oocyte destruction and ovarian failure occur in PAH-treated mice, and cigarette smoking causes early menopause in women. In many cells, PAHs activate the aromatic hydrocarbon receptor (Ahr), a member of the Per-Arnt-Sim family of transcription factors. The Ahr is also activated by dioxin, one of the most intensively studied environmental contaminants. Here we show that an exposure of mice to PAHs induces the expression of Bax in oocytes, followed by apoptosis. Ovarian damage caused by PAHs is prevented by Ahr or Bax inactivation. Oocytes microinjected with a Bax promoter-reporter construct show Ahr-dependent transcriptional activation after PAH, but not dioxin, treatment, consistent with findings that dioxin is not cytotoxic to oocytes. This difference in the action of PAHs versus dioxin is conveyed by a single base pair flanking each Ahr response element in the Bax promoter. Oocytes in human ovarian biopsies grafted into immunodeficient mice also accumulate Bax and undergo apoptosis after PAH exposure in vivo. Thus, Ahr-driven Bax transcription is a novel and evolutionarily conserved cell-death signaling pathway responsible for environmental toxicant-induced ovarian failure.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene/analogs & derivatives
- Adult
- Animals
- Apoptosis
- Environmental Pollution/adverse effects
- Female
- Gene Expression/drug effects
- Genes, Reporter
- Humans
- In Vitro Techniques
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Microinjections
- Oocytes/cytology
- Oocytes/drug effects
- Oocytes/metabolism
- Ovary/drug effects
- Ovary/metabolism
- Ovary/transplantation
- Primary Ovarian Insufficiency/chemically induced
- Primary Ovarian Insufficiency/genetics
- Promoter Regions, Genetic
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins c-bcl-2
- Receptors, Aryl Hydrocarbon/deficiency
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Response Elements
- Signal Transduction/drug effects
- Transplantation, Heterologous
- bcl-2-Associated X Protein
Collapse
Affiliation(s)
- T Matikainen
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Black AR, Black JD, Azizkhan-Clifford J. Sp1 and krüppel-like factor family of transcription factors in cell growth regulation and cancer. J Cell Physiol 2001; 188:143-60. [PMID: 11424081 DOI: 10.1002/jcp.1111] [Citation(s) in RCA: 830] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Sp/KLF family contains at least twenty identified members which include Sp1-4 and numerous krüppel-like factors. Members of the family bind with varying affinities to sequences designated as 'Sp1 sites' (e.g., GC-boxes, CACCC-boxes, and basic transcription elements). Family members have different transcriptional properties and can modulate each other's activity by a variety of mechanisms. Since cells can express multiple family members, Sp/KLF factors are likely to make up a transcriptional network through which gene expression can be fine-tuned. 'Sp1 site'-dependent transcription can be growth-regulated, and the activity, expression, and/or post-translational modification of multiple family members is altered with cell growth. Furthermore, Sp/KLF factors are involved in many growth-related signal transduction pathways and their overexpression can have positive or negative effects on proliferation. In addition to growth control, Sp/KLF factors have been implicated in apoptosis and angiogenesis; thus, the family is involved in several aspects of tumorigenesis. Consistent with a role in cancer, Sp/KLF factors interact with oncogenes and tumor suppressors, they can be oncogenic themselves, and altered expression of family members has been detected in tumors. Effects of changes in Sp/KLF factors are context-dependent and can appear contradictory. Since these factors act within a network, this diversity of effects may arise from differences in the expression profile of family members in various cells. Thus, it is likely that the properties of the overall network of Sp/KLF factors play a determining role in regulation of cell growth and tumor progression.
Collapse
Affiliation(s)
- A R Black
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA.
| | | | | |
Collapse
|
26
|
Cinti C, Claudio PP, Luca AD, Cuccurese M, Howard CM, D'Esposito M, Paggi MG, Sala DL, Azzoni L, Halazonetis TD, Giordano A, Maraldi NM. A serine 37 mutation associated with two missense mutations at highly conserved regions of p53 affect pro-apoptotic genes expression in a T-lymphoblastoid drug resistant cell line. Oncogene 2000; 19:5098-105. [PMID: 11042698 DOI: 10.1038/sj.onc.1203848] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The p53 protein accumulates rapidly through post-transcriptional mechanisms following cellular exposure to DNA damaging agents and is also activated as a transcription factor leading to growth arrest or apoptosis. Phosphorylation of p53 occurs after DNA damage thereby modulating its activity and impeding the interaction of p53 with its negative regulator oncogene Mdm2. The serines 15 and 37 present in the amino terminal region of p53 are phosphorylated by the DNA-dependent protein kinase (DNA-PK) in response to DNA damage. In order to verify if specific p53 mutations occur in the multi-drug resistance phenotype, we analysed the p53 gene in two T-lymphoblastoid cell lines, CCRF-CEM and its multi-drug-resistant clone CCRF-CEM VLB100, selected for resistance to vinblastine sulfate and cross-resistant to other cytotoxic drugs. Both cell lines showed two heterozygous mutations in the DNA binding domain at codons 175 and 248. The multi-drug resistant cell line, CCRF-CEM VLB100, showed an additional mutation that involves the serine 37 whose phosphorylation is important to modulate the protein activity in response to DNA damage. The effects of these mutations on p53 transactivation capacity were evaluated. The activity of p53 on pro-apoptotic genes expression in response to DNA damage induced by (-irradiation, was affected in the vinblastine (VLB) resistant cell line but not in CCRF-CEM sensitive cell line resulting in a much reduced apoptotic cell death of the multi-drug resistant cells.
Collapse
MESH Headings
- Amino Acid Substitution
- Antibiotics, Antineoplastic/pharmacology
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis/genetics
- Base Sequence
- Cell Survival/radiation effects
- Conserved Sequence
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- DNA, Neoplasm/radiation effects
- Dactinomycin/pharmacology
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/genetics
- Drug Resistance, Neoplasm/genetics
- Exons
- Gene Expression Regulation, Leukemic/genetics
- Genes, p53/genetics
- Humans
- Leukemia, T-Cell/genetics
- Leukemia, T-Cell/metabolism
- Leukemia, T-Cell/pathology
- Mutation, Missense
- Polymorphism, Single-Stranded Conformational
- Radiation Tolerance/genetics
- Serine/genetics
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/radiation effects
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/physiology
- Vinblastine/pharmacology
Collapse
Affiliation(s)
- C Cinti
- Institute of Normal and Pathologic Cytomorphology, CNR, c/o IOR, 40136 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|