Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Genes Dev ; 33(5-6): 310-332, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30804224

ABSTRACT

Whether cell types exposed to a high level of environmental insults possess cell type-specific prosurvival mechanisms or enhanced DNA damage repair capacity is not well understood. BRN2 is a tissue-restricted POU domain transcription factor implicated in neural development and several cancers. In melanoma, BRN2 plays a key role in promoting invasion and regulating proliferation. Here we found, surprisingly, that rather than interacting with transcription cofactors, BRN2 is instead associated with DNA damage response proteins and directly binds PARP1 and Ku70/Ku80. Rapid PARP1-dependent BRN2 association with sites of DNA damage facilitates recruitment of Ku80 and reprograms DNA damage repair by promoting Ku-dependent nonhomologous end-joining (NHEJ) at the expense of homologous recombination. BRN2 also suppresses an apoptosis-associated gene expression program to protect against UVB-, chemotherapy- and vemurafenib-induced apoptosis. Remarkably, BRN2 expression also correlates with a high single-nucleotide variation prevalence in human melanomas. By promoting error-prone DNA damage repair via NHEJ and suppressing apoptosis of damaged cells, our results suggest that BRN2 contributes to the generation of melanomas with a high mutation burden. Our findings highlight a novel role for a key transcription factor in reprogramming DNA damage repair and suggest that BRN2 may impact the response to DNA-damaging agents in BRN2-expressing cancers.


Subject(s)
Apoptosis , DNA End-Joining Repair/genetics , Homeodomain Proteins/metabolism , Melanoma/genetics , Melanoma/physiopathology , Mutation/genetics , POU Domain Factors/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Humans , Ku Autoantigen/metabolism , POU Domain Factors/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding , Protein Domains , Protein Transport
2.
Br J Cancer ; 117(4): 503-512, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28677687

ABSTRACT

BACKGROUND: Tumour-specific radiosensitising treatments may enhance the efficacy of radiotherapy without exacerbating side effects. In this study we determined the radiation response following depletion or inhibition of TOPK, a mitogen-activated protein kinase kinase family Ser/Thr protein kinase that is upregulated in many cancers. METHODS: Radiation response was studied in a wide range of cancer cell lines and normal cells using colony formation assays. The effect on cell cycle progression was assessed and the relationship between TOPK expression and therapeutic efficacy was studied in a cohort of 128 prostate cancer patients treated with radical radiotherapy. RESULTS: TOPK knockdown did not alter radiation response in normal tissues, but significantly enhanced radiosensitivity in cancer cells. This result was recapitulated in TOPK knockout cells and with the TOPK inhibitor, OTS964. TOPK depletion altered the G1/S transition and G2/M arrest in response to radiation. Furthermore, TOPK depletion increased chromosomal aberrations, multinucleation and apoptotic cell death after irradiation. These results suggest a possible role for TOPK in the radiation-induced DNA damage checkpoints. These findings have clinical relevance, as elevated TOPK protein expression was associated with poorer clinical outcomes in prostate cancer patients treated with radical radiotherapy. CONCLUSIONS: This study demonstrates that TOPK disruption may cause tumour-specific radiosensitisation in multiple different tumour types.


Subject(s)
Cell Cycle Checkpoints , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Recurrence, Local/metabolism , Prostatic Neoplasms/radiotherapy , Radiation Tolerance , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/radiation effects , Chromosome Aberrations/drug effects , Chromosome Aberrations/radiation effects , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Male , Mitogen-Activated Protein Kinase Kinases/genetics , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Survival Rate
3.
Support Care Cancer ; 24(5): 2317-2321, 2016 May.
Article in English | MEDLINE | ID: mdl-26610767

ABSTRACT

PURPOSE: Despite substantial morbidity and mortality of influenza and pneumococcal infections in cancer patients treated with chemotherapy, vaccination against both illnesses is infrequent. We evaluated the impact of implementation of clinical guidelines on vaccination of chemotherapy patients treated in our institute. METHODS: We performed a prospective audit before (2012) and after (2013-2014) the introduction of immunisation guidelines for chemotherapy patients in a UK tertiary cancer centre. RESULTS: Guideline implementation was associated with a significant increase in the rate of pneumococcal vaccination compared to the 2012 baseline (47 vs. 25 %, P = 0.0018), though this was not sustained the following year (34 %, P = 0.13, vs. baseline). Influenza vaccine coverage was high (∼ 70 %) throughout. There was a marked disparity between patients aged ≤ 65 and those >65 years in the rate of pneumococcal vaccination in both 2013 and 2014 (38 vs. 68 % and 17 vs. 53 %, respectively, both P < 0.001), and, to a lesser extent, in the rate of influenza vaccination in the same period (64 vs. 82 %, P < 0.1, and 63 vs. 85 %, P = 0.009, respectively). CONCLUSIONS: The implementation of clinical vaccine guidelines was associated with a significant increase in pneumococcal vaccination, though continued effort appears required to deliver persistent improvement. Initiatives to increase vaccination uptake in patients aged ≤ 65 are merited.


Subject(s)
Influenza Vaccines/therapeutic use , Neoplasms/therapy , Vaccination/methods , Adult , Aged , Aged, 80 and over , Clinical Audit , Female , Humans , Influenza Vaccines/administration & dosage , Male , Middle Aged , Pneumococcal Infections , Prospective Studies , Young Adult
4.
Toxicol Appl Pharmacol ; 277(3): 288-97, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24726431

ABSTRACT

Mutations to the p53 gene are common in UV-exposed keratinocytes and contribute to apoptotic resistance in skin cancer. P53-dependent activity is modulated, in part, by a complex, self-limiting feedback loop imposed by miR-34a-mediated regulation of the lysine deacetylase, SIRT1. Expression of numerous microRNAs is dysregulated in squamous and basal cell carcinomas; however the contribution of specific microRNAs to the pathogenesis of skin cancer remains untested. Through use of RNAi, miRNA target site blocking oligonucleotides and small molecule inhibitors, this study explored the influence of p53 mutational status, SIRT1 activity and miR-34a levels on apoptotic sensitivity in primary (NHEK) and p53-mutated (HaCaT) keratinocyte cell lines. SIRT1 and p53 are overexpressed in p53-mutated keratinocytes, whilst miR-34a levels are 90% less in HaCaT cells. HaCaTs have impaired responses to p53/SIRT1/miR-34a axis manipulation which enhanced survival during exposure to the chemotherapeutic agent, camptothecin. Inhibition of SIRT1 activity in this cell line increased p53 acetylation and doubled camptothecin-induced cell death. Our results demonstrate that p53 mutations increase apoptotic resistance in keratinocytes by interfering with miR-34a-mediated regulation of SIRT1 expression. Thus, SIRT1 inhibitors may have a therapeutic potential for overcoming apoptotic resistance during skin cancer treatment.


Subject(s)
Apoptosis/physiology , Keratinocytes/metabolism , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Gene Expression Regulation , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Oligonucleotides , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Survivin , Tumor Suppressor Protein p53/genetics
5.
Toxicol Appl Pharmacol ; 281(1): 136-45, 2014 11 15.
Article in English | MEDLINE | ID: mdl-25281835

ABSTRACT

Arsenic is an environmental toxin which increases skin cancer risk for exposed populations worldwide; however the underlying biomolecular mechanism for arsenic-induced carcinogenesis is complex and poorly defined. Recent investigations show that histone deacetylase and DNA methyltransferase activity is impaired, and epigenetic patterns of gene regulation are consistently altered in cancers associated with arsenic exposure. Expression of the histone deacetylase SIRT1 is altered in solid tumours and haematological malignancies; however its role in arsenic-induced pathology is unknown. In this study we investigated the effect of arsenic on epigenetic regulation of SIRT1 and its targeting microRNA, miR-34a in primary human keratinocytes. Acetylation of histone H4 at lysine 16 (H4K16) increased in keratinocytes exposed to 0.5µM arsenite [As(III)]; and this was associated with chromatin remodelling at the miR-34a promoter. Moreover, although SIRT1 protein initially increased in these As(III)-exposed cells, after 24days expression was not significantly different from untreated controls. Extended exposure to low-dose As(III) (0.5µM; >5weeks) compromised the pattern of CpG methylation at SIRT1 and miR-34a gene promoters, and this was associated with altered expression for both genes. We have found that arsenic alters epigenetic regulation of SIRT1 expression via structural reorganisation of chromatin at the miR-34a gene promoter in the initial 24h of exposure; and over time, through shifts in miR-34a and SIRT1 gene methylation. Taken together, this investigation demonstrates that arsenic produces cumulative disruptions to epigenetic regulation of miR-34a expression, and this is associated with impaired coordination of SIRT1 functional activity.


Subject(s)
Arsenic/toxicity , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/physiology , Keratinocytes/drug effects , Keratinocytes/physiology , Sirtuin 1/biosynthesis , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Sirtuin 1/genetics
6.
Sci Rep ; 14(1): 8998, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38637546

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is considered the third leading cause of cancer mortality in the western world, offering advanced stage patients with few viable treatment options. Consequently, there remains an urgent unmet need to develop novel therapeutic strategies that can effectively inhibit pro-oncogenic molecular targets underpinning PDACs pathogenesis and progression. One such target is c-RAF, a downstream effector of RAS that is considered essential for the oncogenic growth and survival of mutant RAS-driven cancers (including KRASMT PDAC). Herein, we demonstrate how a novel cell-penetrating peptide disruptor (DRx-170) of the c-RAF-PDE8A protein-protein interaction (PPI) represents a differentiated approach to exploiting the c-RAF-cAMP/PKA signaling axes and treating KRAS-c-RAF dependent PDAC. Through disrupting the c-RAF-PDE8A protein complex, DRx-170 promotes the inactivation of c-RAF through an allosteric mechanism, dependent upon inactivating PKA phosphorylation. DRx-170 inhibits cell proliferation, adhesion and migration of a KRASMT PDAC cell line (PANC1), independent of ERK1/2 activity. Moreover, combining DRx-170 with afatinib significantly enhances PANC1 growth inhibition in both 2D and 3D cellular models. DRx-170 sensitivity appears to correlate with c-RAF dependency. This proof-of-concept study supports the development of DRx-170 as a novel and differentiated strategy for targeting c-RAF activity in KRAS-c-RAF dependent PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Signal Transduction , Cell Proliferation , Cell Line, Tumor , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism
7.
Lancet ; 379(9830): 1975-82, 2012 May 26.
Article in English | MEDLINE | ID: mdl-22521520

ABSTRACT

BACKGROUND: The burden of non-communicable diseases (NCDs) is disproportionately carried by low-income and middle-income countries and disadvantaged sectors of society such as prisoners. No systematic analysis has been done to assess the prevalence of poor diet, inadequate physical activity, and overweight and obesity in prisoners. We aim to synthesise current evidence and to highlight areas for action and further research. METHODS: We systematically searched online databases for reports published between 1948 and May, 2011. Studies were screened against eligibility criteria; two authors then independently extracted data with previously agreed proformas. The risk of bias was assessed for each study with a domain-based assessment. Data on body-mass index and physical activity were presented in forest plots; no overall estimates were calculated on account of data heterogeneity. Available data from the population subgroup most similar in terms of age and sex were used to calculate age-adjusted and sex-adjusted prevalence ratios, which estimate the likelihood of insufficient activity and obesity prevalence in prisoners compared with the national population. FINDINGS: 31 eligible studies were reported in 29 publications, including more than 60,000 prisoners in 884 institutions in 15 countries. Male prisoners were less likely to be obese than males in the general population (prevalence ratios ranged from 0·33 to 0·87) in all but one study (1·02, 0·92-1·07), whereas female prisoners were more likely to be obese than non-imprisoned women in the USA (1·18, 1·08-1·30) and Australia (prevalence ratios ranged from 1·15 to 1·20). Australian prisoners were more likely to achieve sufficient activity levels than the general population compared with prisoners in the UK (prevalence ratio 1·19, 95% CI 1·04-1·37, for women in Australia in 2009 vs 0·32, 0·21-0·47, for women in the UK; prevalence ratios ranged from 1·37 to 1·59 for men in Australia vs 0·71, 0·34-0·78, for men in the UK). Female mean energy intake exceeded recommended levels and sodium intake was about two to three times the recommended intake for all prisoners. INTERPRETATION: Contact with the criminal justice system is a public-health opportunity to promote health in this vulnerable population; the costs to the individual and to society of failing to do so are likely to be substantial. Improved monitoring and further research is essential to inform appropriate targeting of public health interventions. FUNDING: Oxford University Department of Public Health, Oxford University Hospitals NHS Trust.


Subject(s)
Diet/statistics & numerical data , Exercise , Obesity/epidemiology , Prisoners/statistics & numerical data , Adult , Body Mass Index , Energy Intake , Female , Humans , Male , Overweight/epidemiology , Prevalence , Risk Factors , Young Adult
8.
Cell Death Differ ; 28(4): 1333-1346, 2021 04.
Article in English | MEDLINE | ID: mdl-33168956

ABSTRACT

T-LAK-originated protein kinase (TOPK) overexpression is a feature of multiple cancers, yet is absent from most phenotypically normal tissues. As such, TOPK expression profiling and the development of TOPK-targeting pharmaceutical agents have raised hopes for its future potential in the development of targeted therapeutics. Results presented in this paper confirm the value of TOPK as a potential target for the treatment of solid tumours, and demonstrate the efficacy of a TOPK inhibitor (OTS964) when used in combination with radiation treatment. Using H460 and Calu-6 lung cancer xenograft models, we show that pharmaceutical inhibition of TOPK potentiates the efficacy of fractionated irradiation. Furthermore, we provide in vitro evidence that TOPK plays a hitherto unknown role during S phase, showing that TOPK depletion increases fork stalling and collapse under conditions of replication stress and exogenous DNA damage. Transient knockdown of TOPK was shown to impair recovery from fork stalling and to increase the formation of replication-associated single-stranded DNA foci in H460 lung cancer cells. We also show that TOPK interacts directly with CHK1 and Cdc25c, two key players in the checkpoint signalling pathway activated after replication fork collapse. This study thus provides novel insights into the mechanism by which TOPK activity supports the survival of cancer cells, facilitating checkpoint signalling in response to replication stress and DNA damage.


Subject(s)
Checkpoint Kinase 1/drug effects , Lung Neoplasms/radiotherapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Radiation Tolerance/drug effects , cdc25 Phosphatases/drug effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/radiation effects , Female , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Radiation Tolerance/genetics , Signal Transduction , Survival Rate , Xenograft Model Antitumor Assays , cdc25 Phosphatases/genetics , cdc25 Phosphatases/radiation effects
9.
Nat Commun ; 12(1): 3318, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083536

ABSTRACT

Dormancy, a reversible quiescent cellular state characterized by greatly reduced metabolic activity, protects from genetic damage, prolongs survival and is crucial for tissue homeostasis and cellular response to injury or transplantation. Dormant cells have been characterized in many tissues, but their identification, isolation and characterization irrespective of tissue of origin remains elusive. Here, we develop a live cell ratiometric fluorescent Optical Stem Cell Activity Reporter (OSCAR) based on the observation that phosphorylation of RNA Polymerase II (RNApII), a hallmark of active mRNA transcription elongation, is largely absent in dormant stem cells from multiple lineages. Using the small intestinal crypt as a model, OSCAR reveals in real time the dynamics of dormancy induction and cellular differentiation in vitro, and allows the identification and isolation of several populations of transcriptionally diverse OSCARhigh and OSCARlow intestinal epithelial cell states in vivo. In particular, this reporter is able to identify a dormant OSCARhigh cell population in the small intestine. OSCAR therefore provides a tool for a better understanding of dormant stem cell biology.


Subject(s)
RNA Polymerase II/metabolism , Resting Phase, Cell Cycle/physiology , Animals , Cell Separation , Cyclin-Dependent Kinase 9/metabolism , Flow Cytometry , Fluorescent Dyes/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Transcription, Genetic
10.
Cancers (Basel) ; 12(4)2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32224979

ABSTRACT

Ionising radiation causes cell death through the induction of DNA damage, particularly double-stranded DNA (dsDNA) breaks. Evidence suggests that adenoviruses inhibit proteins involved in the DNA damage response (DDR) to prevent recognition of double-stranded viral DNA genomes as cellular dsDNA breaks. We hypothesise that combining adenovirus treatment with radiotherapy has the potential for enhancing tumour-specific cytotoxicity through inhibition of the DDR and augmentation of virus production. We show that EnAd, an Ad3/Ad11p chimeric oncolytic adenovirus currently being trialled in colorectal and other cancers, targets the DDR pathway at a number of junctures. Infection is associated with a decrease in irradiation-induced 53BP1 and Rad51 foci formation, and in total DNA ligase IV levels. We also demonstrate a radiation-associated increase in EnAd production in vitro and in a pilot in vivo experiment. Given the current limitations of in vitro techniques in assessing for synergy between these treatments, we adapted the plaque assay to allow monitoring of viral plaque size and growth and utilised the xCELLigence cell adhesion assay to measure cytotoxicity. Our study provides further evidence on the interaction between adenovirus and radiation in vitro and in vivo and suggests these have at least an additive, and possibly a synergistic, impact on cytotoxicity.

11.
Cell Death Dis ; 9(11): 1089, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356039

ABSTRACT

'Targeted' or 'biological' cancer treatments rely on differential gene expression between normal tissue and cancer, and genetic changes that render tumour cells especially sensitive to the agent being applied. Problems exist with the application of many agents as a result of damage to local tissues, tumour evolution and treatment resistance, or through systemic toxicity. Hence, there is a therapeutic need to uncover specific clinical targets which enhance the efficacy of cancer treatment whilst minimising the risk to healthy tissues. T-LAK cell-originated protein kinase (TOPK) is a MAPKK-like kinase which plays a role in cell cycle regulation and mitotic progression. As a consequence, TOPK expression is minimal in differentiated cells, although its overexpression is a pathophysiological feature of many tumours. Hence, TOPK has garnered interest as a cancer-specific biomarker and biochemical target with the potential to enhance cancer therapy whilst causing minimal harm to normal tissues. Small molecule inhibitors of TOPK have produced encouraging results as a stand-alone treatment in vitro and in vivo, and are expected to advance into clinical trials in the near future. In this review, we present the current literature pertaining to TOPK as a potential clinical target and describe the progress made in uncovering its role in tumour development. Firstly, we describe the functional role of TOPK as a pro-oncogenic kinase, followed by a discussion of its potential as a target for the treatment of cancers with high-TOPK expression. Next, we provide an overview of the current preclinical progress in TOPK inhibitor discovery and development, with respect to future adaptation for clinical use.


Subject(s)
Indolizines/therapeutic use , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Quinolones/therapeutic use , Quinoxalines/therapeutic use , Thiophenes/therapeutic use , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Discovery/methods , Humans , Indolizines/pharmacology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Quinolones/pharmacology , Quinoxalines/pharmacology , Thiophenes/pharmacology , Treatment Outcome
12.
Cell Cycle ; 17(12): 1513-1523, 2018.
Article in English | MEDLINE | ID: mdl-30045664

ABSTRACT

Cyclin-dependent kinase 1 (CDK1) orchestrates the transition from the G2 phase into mitosis and as cancer cells often display enhanced CDK1 activity, it has been proposed as a tumor specific anti-cancer target. Here we show that the effects of CDK1 inhibition are not restricted to tumor cells but can also reduce viability in non-cancer cells and sensitize them to radiation in a cell cycle dependent manner. Radiosensitization by the specific CDK1 inhibitor, RO-3306, was determined by colony formation assays in three tumor lines (HeLa, T24, SQ20B) and three non-cancer lines (HFL1, MRC-5, RPE). Initial results showed that CDK1 inhibition radiosensitized tumor cells, but did not sensitize normal fibroblasts and epithelial cells in colony formation assays despite effective inhibition of CDK1 signaling. Further investigation showed that normal cells were less sensitive to CDK1 inhibition because they remained predominantly in G1 for a prolonged period when plated in colony formation assays. In contrast, inhibiting CDK1 a day after plating, when the cells were going through G2/M phase, reduced their clonogenic survival both with and without radiation. Our finding that inhibition of CDK1 can damage normal cells in a cell cycle dependent manner indicates that targeting CDK1 in cancer patients may lead to toxicity in normal proliferating cells. Furthermore, our finding that cell cycle progression becomes easily stalled in non-cancer cells under normal culture conditions has general implications for testing anti-cancer agents in these cells.


Subject(s)
CDC2 Protein Kinase/antagonists & inhibitors , Cell Division/drug effects , DNA Damage/drug effects , G2 Phase/drug effects , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , HeLa Cells , Humans , Signal Transduction/drug effects
13.
J Dermatol Sci ; 74(2): 142-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24548601

ABSTRACT

BACKGROUND: Together with p53, the NAD-dependent lysine deacetylase SIRT1 and the microRNA miR-34a form a feedback loop which self-regulates SIRT1 expression and modulates p53-dependent responses. In addition to its well-described role in mediating transcriptional responses to genotoxic stress, p53 may also regulate microRNA processing and maturation. OBJECTIVE: This study explored the functional relationship among p53, SIRT1 and miR-34a, and the influence of p53 and SIRT1 on microRNA biogenesis and maturation in primary (NHEK) and p53-mutated (HaCaT) keratinocyte cell lines. METHODS: RNAi, miRNA target site blocking oligonucleotides and small molecule inhibitors were used to modulate activity and expression of SIRT1 and p53. Changes in microRNA and mRNA were analysed by qRT-PCR and protein expression was determined by immunoblotting. RESULTS: Mature miR-34a decreased in p53-suppressed NHEK cells, whereas ablation of SIRT1 reduced the primary transcript (pri-miR-34a). When either SIRT1 expression or activity was inhibited in combination with p53 ablation, pri-miR-34a levels increased and mature miR-34a levels decreased. Under these same conditions, additional p53-regulated microRNAs (miRs 16-1/15, 145 and 107) also failed to mature. In HaCaT cells, primary microRNA transcripts for miR-16-1/15, miR-145 miR200c/141 and miRNA-107, but not miR-34a, were approximately 8-fold higher than in NHEK cells. However, the levels of mature microRNA sequences in HaCaT cells were only 1.5-2 fold higher (miR-16-1, miR-145), unchanged (miR-107) or decreased (miR-200c/141, miR-34a) compared to NHEK cells. CONCLUSIONS: Our results suggest that p53 mutations interfere with efficient microRNA biogenesis in keratinocytes, and that SIRT1 functions in combination with p53 in this process.


Subject(s)
Genes, p53 , Keratinocytes/metabolism , MicroRNAs/biosynthesis , Sirtuin 1/metabolism , Cells, Cultured , Humans , MicroRNAs/metabolism
14.
Chem Biol Interact ; 198(1-3): 38-48, 2012 Jun 25.
Article in English | MEDLINE | ID: mdl-22634503

ABSTRACT

Arsenic (As) is both a human carcinogen and an effective anticancer drug. These aspects of arsenic toxicity develop as a consequence of arsenic-induced oxidative stress and modifications to signal pathway activity which alter gene expression. Resveratrol (RVL) a food antioxidant found in grapes and other fruits, exhibits anti-carcinogenic properties by reducing oxidative stress and restoring signal pathway control. This study investigated the impact of RVL on arsenite [As(III)]-induced cell signalling in HaCaT keratinocytes by assaying phosphorylation status of epidermal growth factor receptor (EGFR) signalling intermediates and measuring changes in expression of Phase II and DNA repair biomarkers. As(III) exposure produced dose-dependent toxicity which was associated with increased activation of EGFR pathway intermediates, cSrc, Rac1 and extracellular signal-regulated kinases 1 and 2 (ERK1/2). Arsenic-mediated ERK1/2 activation negatively regulated DNA polymerase beta expression and up regulated heme-oxygenase-1 at toxic concentrations. RVL treatment modulated As(III)-mediated ERK1/2 activation by shifting the balance of cSrc regulatory domain phosphorylation. These effects significantly altered the response of the EGFR pathway to growth factor-induced stimulation. Our research provides evidence that treatment with pharmacologically relevant doses of RVL influences cellular responses to As(III), largely due to RVL-mediated changes to Src and ERK1/2 activation.


Subject(s)
Arsenic/pharmacology , ErbB Receptors/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Base Sequence , Blotting, Western , Cell Line, Transformed , DNA Primers , Dose-Response Relationship, Drug , Humans , Phosphorylation , Real-Time Polymerase Chain Reaction , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL