Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Int J Mol Sci ; 24(4)2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36835644

ABSTRACT

In vivo models of acute myeloid leukemia (AML) are low throughput, and standard liquid culture models fail to recapitulate the mechanical and biochemical properties of the extracellular matrix-rich protective bone marrow niche that contributes to drug resistance. Candidate drug discovery in AML requires advanced synthetic platforms to improve our understanding of the impact of mechanical cues on drug sensitivity in AML. By use of a synthetic, self-assembling peptide hydrogel (SAPH) of modifiable stiffness and composition, a 3D model of the bone marrow niche to screen repurposed FDA-approved drugs has been developed and utilized. AML cell proliferation was dependent on SAPH stiffness, which was optimized to facilitate colony growth. Three candidate FDA-approved drugs were initially screened against the THP-1 cell line and mAF9 primary cells in liquid culture, and EC50 values were used to inform drug sensitivity assays in the peptide hydrogel models. Salinomycin demonstrated efficacy in both an 'early-stage' model in which treatment was added shortly after initiation of AML cell encapsulation, and an 'established' model in which time-encapsulated cells had started to form colonies. Sensitivity to Vidofludimus treatment was not observed in the hydrogel models, and Atorvastatin demonstrated increased sensitivity in the 'established' compared to the 'early-stage' model. AML patient samples were equally sensitive to Salinomycin in the 3D hydrogels and partially sensitive to Atorvastatin. Together, this confirms that AML cell sensitivity is drug- and context-specific and that advanced synthetic platforms for higher throughput are valuable tools for pre-clinical evaluation of candidate anti-AML drugs.


Subject(s)
Hydrogels , Leukemia, Myeloid, Acute , Humans , Hydrogels/therapeutic use , Atorvastatin/therapeutic use , Leukemia, Myeloid, Acute/metabolism , Bone Marrow/metabolism , Peptides/therapeutic use
2.
BMC Cancer ; 20(1): 629, 2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32635894

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is a heterogenous hematological malignancy with poor long-term survival. New drugs which improve the outcome of AML patients are urgently required. In this work, the activity and mechanism of action of the cytotoxic indole alkaloid Jerantinine B (JB), was examined in AML cells. METHODS: We used a combination of proliferation and apoptosis assays to assess the effect of JB on AML cell lines and patient samples, with BH3 profiling being performed to identify early effects of the drug (4 h). Phosphokinase arrays were adopted to identify potential driver proteins in the cellular response to JB, the results of which were confirmed and extended using western blotting and inhibitor assays and measuring levels of reactive oxygen species. RESULTS: AML cell growth was significantly impaired following JB exposure in a dose-dependent manner; potent colony inhibition of primary patient cells was also observed. An apoptotic mode of death was demonstrated using Annexin V and upregulation of apoptotic biomarkers (active caspase 3 and cleaved PARP). Using BH3 profiling, JB was shown to prime cells to apoptosis at an early time point (4 h) and phospho-kinase arrays demonstrated this to be associated with a strong upregulation and activation of both total and phosphorylated c-Jun (S63). The mechanism of c-Jun activation was probed and significant induction of reactive oxygen species (ROS) was demonstrated which resulted in an increase in the DNA damage response marker ƎĀ³H2AX. This was further verified by the loss of JB-induced C-Jun activation and maintenance of cell viability when using the ROS scavenger N-acetyl-L-cysteine (NAC). CONCLUSIONS: This work provides the first evidence of cytotoxicity of JB against AML cells and identifies ROS-induced c-Jun activation as the major mechanism of action.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Indole Alkaloids/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins c-jun/agonists , Acetylcysteine/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/drug effects , Drug Screening Assays, Antitumor , Free Radical Scavengers/pharmacology , Humans , Indole Alkaloids/therapeutic use , Leukemia, Myeloid, Acute/pathology , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
3.
Int J Mol Sci ; 20(11)2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31167387

ABSTRACT

Acute myeloid leukaemia (AML) is a heterogeneous clonal malignancy of hematopoietic progenitor cells. The Wnt pathway and its downstream targets are tightly regulated by Ɵ-catenin. We recently discovered a new protein, FLYWCH1, which can directly bind nuclear Ɵ-catenin. Herein, we studied the FLYWCH1/Ɵ-catenin pathway in AML cells using qRT-PCR, Western blot, and immunofluorescence assays. In addition, the stemness activity and cell cycle were analysed by the colony-forming unit (CFU) using methylcellulose-based and Propidium iodide/flow cytometry assays. We found that FLYWCH1 mRNA and protein were differentially expressed in the AML cell lines. C-Myc, cyclin D1, and c-Jun expression decreased in the presence of higher FLYWCH1 expression, and vice versa. There appeared to be the loss of FLYWCH1 expression in dividing cells. The sub-G0 phase was prolonged and shortened in the low and high FLYWCH1 expression cell lines, respectively. The G0/G1 arrest correlated with FLYWCH1-expression, and these cell lines also formed colonies, whereas the low FLYWCH1 expression cell lines could not. Thus, FLYWCH1 functions as a negative regulator of the Wnt/Ɵ-catenin pathway in AML.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Cell Cycle/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Humans , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , Wnt Signaling Pathway
4.
Acta Obstet Gynecol Scand ; 96(5): 556-562, 2017 May.
Article in English | MEDLINE | ID: mdl-28176325

ABSTRACT

INTRODUCTION: Women with polycystic ovary syndrome have a three-fold higher risk of endometrial cancer. Insulin resistance and hyperlipidemia may be pertinent factors in the pathogenesis of both conditions. The aim of this study was to investigate endometrial sterol regulatory element binding protein-1 gene expression in polycystic ovary syndrome and endometrial cancer endometrium, and to correlate endometrial sterol regulatory element binding protein-1 gene expression with serum lipid profiles. MATERIAL AND METHODS: A cross-sectional study was performed at Nottingham University Hospital, UK. A total of 102 women (polycystic ovary syndrome, endometrial cancer and controls; 34 participants in each group) were recruited. Clinical and biochemical assessments were performed before endometrial biopsies were obtained from all participants. Taqman real-time polymerase chain reaction for endometrial sterol regulatory element binding protein-1 gene and its systemic protein expression were analyzed. RESULTS: The body mass indices of women with polycystic ovary syndrome (29.28Ā Ā±Ā 2.91Ā kg/m2 ) and controls (28.58Ā Ā±Ā 2.62Ā kg/m2 ) were not significantly different. Women with endometrial cancer had a higher mean body mass index (32.22Ā Ā±Ā 5.70Ā kg/m2 ). Sterol regulatory element binding protein-1 gene expression was significantly increased in polycystic ovary syndrome and endometrial cancer endometrium compared with controls (pĀ <Ā 0.0001). Sterol regulatory element binding protein-1 gene expression was positively correlated with body mass index (rĀ =Ā 0.017, pĀ =Ā 0.921) and waist-hip ratio (rĀ =Ā 0.023, pĀ =Ā 0.544) in polycystic ovary syndrome, but this was not statistically significant. Similarly, statistically insignificant positive correlations were found between endometrial sterol regulatory element binding protein-1 gene expression and body mass index in endometrial cancer (rĀ =Ā 0.643, pĀ =Ā 0.06) and waist-hip ratio (rĀ =Ā 0.096, pĀ =Ā 0.073). Sterol regulatory element binding protein-1 gene expression was significantly positively correlated with triglyceride in both polycystic ovary syndrome and endometrial cancer (pĀ =Ā 0.028 and pĀ =Ā 0.027, respectively). Quantitative serum sterol regulatory element binding protein-1 gene correlated with endometrial gene expression (pĀ <Ā 0.05). CONCLUSIONS: Sterol regulatory element binding protein-1 gene expression is significantly increased in the endometrium of women with polycystic ovary syndrome and women with endometrial cancer compared with controls and positively correlates with serum triglyceride in both polycystic ovary syndrome and endometrial cancer.


Subject(s)
Endometrial Neoplasms/genetics , Polycystic Ovary Syndrome/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Adult , Case-Control Studies , Cross-Sectional Studies , Endometrial Neoplasms/blood , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic , Humans , Lipids/blood , Middle Aged , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
5.
J Immunol ; 190(7): 3246-55, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460736

ABSTRACT

Little is known of the regulation of IL-23 secretion in dendritic cells (DC) despite its importance for human Th17 responses. In this study, we show for first time, to our knowledge, that the ataxia telangiectasia mutated (ATM) pathway, involved in DNA damage sensing, acts as an IL-23 repressor. Inhibition of ATM with the highly selective antagonist KU55933 markedly increased IL-23 secretion in human monocyte-derived DC and freshly isolated myeloid DC. In contrast, inhibiting the closely related mammalian target of rapamycin had no effect on IL-23. Priming naive CD4(+) T cells with ATM-inhibited DC increased Th17 responses over and above those obtained with mature DC. Although ATM blockade increased the abundance of p19, p35, and p40 mRNA, IL-12p70 secretion was unaffected. To further examine a role for ATM in IL-23 regulation, we exposed DC to low doses of ionizing radiation. Exposure of DC to x-rays resulted in ATM phosphorylation and a corresponding depression of IL-23. Importantly, ATM inhibition with KU55933 prevented radiation-induced ATM phosphorylation and abrogated the capacity of x-rays to suppress IL-23. To explore how ATM repressed IL-23, we examined a role for endoplasmic reticulum stress responses by measuring generation of the spliced form of X-box protein-1, a key endoplasmic reticulum stress transcription factor. Inhibition of ATM increased the abundance of X-box protein-1 mRNA, and this was followed 3 h later by increased peak p19 transcription and IL-23 release. In summary, ATM activation or inhibition, respectively, inhibited or augmented IL-23 release. This novel role of the ATM pathway represents a new therapeutic target in autoimmunity and vaccine development.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Gene Expression Regulation , Interleukin-23/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Endoplasmic Reticulum Stress , Enzyme Activation/radiation effects , Gene Expression Regulation/radiation effects , Humans , Interleukin-23/metabolism , Lymphocyte Activation/immunology , Regulatory Factor X Transcription Factors , Signal Transduction/radiation effects , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
6.
Gynecol Endocrinol ; 31(4): 286-90, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25495168

ABSTRACT

The aim of this proof-of-concept study was to determine the effects of three-month Metformin therapy on the expression of tumor-regulatory genes (p53, cyclin D2 and BCL-2) in the endometrium of women with polycystic ovary syndrome (PCOS). A total of 40 women, aged between 21 and 45 years with PCOS (Rotterdam criteria) were recruited. The participants were assessed at pre- and 3-month-post-Metformin therapy for the menstrual regularities, weight reduction, Ferriman Galway scores, fasting blood glucose (FBG), total cholesterol, LDL, HDL and p53, BCL-2 and cyclin D2 gene expression. Five participants conceived spontaneously after the initial recruitment. Majority (68%) resumed regular menstrual cycles after Metformin. There were significant reduction in BMI (p = 0.001), weight (p = 0.001) and Ferriman Galway scores (p = 0.001). A significant improvement was seen in mean FBG (p = 0.002), total cholesterol (p = 0.001), LDL (p = 0.003) and HDL cholesterol levels (p = 0.015). Tumor suppressor gene (p53) was significantly up-regulated after Metformin (10 out of 14 women), with p value 0.016. BCL-2 and cyclin D2 (oncogenes) were slightly up-regulated without significant difference (p = 0.119 and 0.155, respectively). In conclusion, Metformin therapy improved clinical and metabolic parameters in women with PCOS and up-regulated p53 tumor suppressor gene significantly. Further studies are however required to independently validate our findings.


Subject(s)
Endometrium/drug effects , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Overweight/prevention & control , Polycystic Ovary Syndrome/drug therapy , Tumor Suppressor Protein p53/agonists , Up-Regulation/drug effects , Adult , Biopsy , Body Mass Index , Cohort Studies , Cyclin D2/agonists , Cyclin D2/genetics , Cyclin D2/metabolism , Endometrial Neoplasms/complications , Endometrial Neoplasms/epidemiology , Endometrial Neoplasms/prevention & control , Endometrium/metabolism , Endometrium/pathology , Female , Follicular Phase/metabolism , Humans , Hypoglycemic Agents/adverse effects , Malaysia/epidemiology , Metformin/adverse effects , Overweight/complications , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Prospective Studies , Proto-Oncogene Proteins c-bcl-2/agonists , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Risk , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Weight Loss/drug effects , Young Adult
8.
Gynecol Oncol ; 132(1): 248-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24183733

ABSTRACT

Current data indicate that there is a significant risk of endometrial cancer (EC) in women with polycystic ovarian syndrome (PCOS), although further research needed to clarify the exact molecular mechanisms. Endometrial hyperplasia is a premalignant condition that usually heralds EC and it shares identical risk factors with EC. Metabolic syndrome with a triad of obesity, hyperinsulinaemia and diabetes, which is commonly observed in PCOS appears to be a key mechanism in EC pathogenesis. Measures to improve insulin resistance could therefore play a role in reducing the risk of EC in women with PCOS. Metformin is an insulin sensitising agent which is safe, widely available and currently licensed for type-2 diabetes. It has been clearly shown in both animal and human studies that metformin is of value in reversing endometrial hyperplasia. Metformin may therefore prevent EC in PCOS. This article reviews the use of metformin in reducing EC risk in PCOS and makes a case for future research on this topic.


Subject(s)
Endometrial Neoplasms/prevention & control , Metformin/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Endometrial Hyperplasia/complications , Female , Humans , Polycystic Ovary Syndrome/complications , Progesterone/therapeutic use , Risk
9.
Int J Cancer ; 132(12): 2778-86, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23225521

ABSTRACT

X-ray repair cross-complementing gene 1 (XRCC1) is essential for DNA base excision repair, single strand break repair and nucleotide excision repair. We investigated clinicopathological and functional significance of XRCC1 expression in ovarian cancers. XRCC1 protein expression was evaluated in 195 consecutive human ovarian cancers and correlated with clinicopathological variables and survival outcomes. Functional preclinical studies were conducted in a panel of XRCC1 deficient and proficient Chinese hamster and Human cancer cells for cisplatin chemosensitivity. Clonogenic assay, neutral COMET assay, ƎĀ³H2AX immunocytochemistry and flow cytometric analyses were performed in cells. In ovarian cancer, 48% of the tumors were positive for XRCC1 expression and significantly associated with higher stage (p = 0.006), serous type tumors (p = 0.008), suboptimal de-bulking (p = 0.004) and platinum resistance (p < 0.0001). Positive XRCC1 had twofold increase of risk of death (p = 0.007) and progression (p < 0.0001). In the multivariate Cox model, XRCC1 expression was independently associated with cancer specific [p = 0.038] and progression free survival [p = 0.003]. Preclinically, XRCC1 negative cells were sensitive to cisplatin compared to XRCC1 positive cells. Sensitivity to cisplatin in XRCC1 negative cells was associated with accumulation of DNA double strand breaks and G2/M cell cycle arrest. XRCC1 expression is associated with adverse clinicopathological and survival outcomes in patients. Preclinical data provides mechanistic functional evidence for cisplatin sensitivity in XRCC1 negative cells. XRCC1 is a promising predictive biomarker in ovarian cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Cell Line , Cricetinae , DNA-Binding Proteins/genetics , Disease Progression , Female , Gene Expression , Gene Silencing , Humans , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Platinum/therapeutic use , RNA Interference , Treatment Outcome , Tumor Stem Cell Assay , X-ray Repair Cross Complementing Protein 1
10.
Int J Cancer ; 131(10): 2433-44, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22377908

ABSTRACT

An apurinic/apyrimidinic (AP) site is an obligatory cytotoxic intermediate in DNA Base Excision Repair (BER) that is processed by human AP endonuclease 1 (APE1). APE1 is essential for BER and an emerging drug target in cancer. We have isolated novel small molecule inhibitors of APE1. In this study, we have investigated the ability of APE1 inhibitors to induce synthetic lethality (SL) in a panel of DNA double-strand break (DSB) repair deficient and proficient cells; i) Chinese hamster (CH) cells: BRCA2 deficient (V-C8), ATM deficient (V-E5), wild type (V79) and BRCA2 revertant [V-C8(Rev1)]. ii) Human cancer cells: BRCA1 deficient (MDA-MB-436), BRCA1 proficient (MCF-7), BRCA2 deficient (CAPAN-1 and HeLa SilenciX cells), BRCA2 proficient (PANC1 and control SilenciX cells). We also tested SL in CH ovary cells expressing a dominant-negative form of APE1 (E8 cells) using ATM inhibitors and DNA-PKcs inhibitors (DSB inhibitors). APE1 inhibitors are synthetically lethal in BRCA and ATM deficient cells. APE1 inhibition resulted in accumulation of DNA DSBs and G2/M cell cycle arrest. SL was also demonstrated in CH cells expressing a dominant-negative form of APE1 treated with ATM or DNA-PKcs inhibitors. We conclude that APE1 is a promising SL target in cancer.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Animals , BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cricetinae , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Humans
11.
Br J Haematol ; 159(2): 191-203, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22934750

ABSTRACT

The novel multi-kinase inhibitor TG02 has selectivity against cell cycle and transcriptional cyclin dependent kinases (CDKs) as well as fms-like tyrosine kinase receptor-3 (FLT3). Inhibition of transcriptional CDKs preferentially depletes short-lived proteins such as MCL1. We evaluated the in vitro toxicity of TG02 to primary acute myeloid leukaemia (AML) cells in the presence of survival signalling pathway activation by cytokines and fibronectin. One hundred nanomolar TG02 induced a median decrease of 40% in bulk cell survival and 43% in the CD34(+) CD38(-) CD123(+) subset. A 90% inhibitory concentration of 500Ā nmol/l indicated that TG02 toxicity is not halted by protective cell cycle arrest. Samples with FLT3 internal tandem duplication were not preferentially targeted. By flow cytometry, TG02 treatment caused loss of RNA Polymerase II serine 2 phosphorylation in patient samples, which correlated strongly with BAX activation (R(2) =0Ā·89), suggesting these as potential biomarkers for clinical studies. MCL1 and XIAP expression also decreased. Repeated brief exposure to TG02 in MOLM-13 cells did not result in compensatory up-regulation of survival protein expression. In conclusion, TG02 is potently cytotoxic towards CD34(+) CD38(-) CD123(+) and bulk AML cells, despite protective signalling pathway activation. This antitumour activity is most likely mediated by dephosphorylation of RNA Polymerase II leading to depletion of survival molecules such as MCL1 and XIAP, with subsequent BAX activation and apoptosis.


Subject(s)
Apoptosis/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism , Antigens, CD , Drug Screening Assays, Antitumor , Female , HL-60 Cells , Humans , Leukemia, Myeloid, Acute , Male , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects , RNA Polymerase II/metabolism , U937 Cells , bcl-2-Associated X Protein/metabolism
12.
BMC Cancer ; 12: 431, 2012 Sep 26.
Article in English | MEDLINE | ID: mdl-23013471

ABSTRACT

BACKGROUND: The CD34+CD38- subset of AML cells is enriched for resistance to current chemotherapeutic agents and considered to contribute to disease progression and relapse in Acute Myeloid Leukaemia (AML) patients following initial treatment. METHODS: Chemosensitivity in phenotypically defined subsets from 34 primary AML samples was measured by flow cytometry following 48 hr in vitro treatment with gemtuzumab ozogamicin (GO, Mylotarg) and the farnesyltransferase inhibitor tipifarnib/zarnestra. The DNA damage response was measured using flow cytometry, immunofluorescence and immunohistochemistry. RESULTS: Using a previously validated in vitro minimal residual disease model, we now show that the combination of GO (10 ng/ml) and tipifarnib (5 ĀµM) targets the CD34+CD38- subset resulting in 65% median cell loss compared to 28% and 13% CD34+CD38- cell loss in GO-treated and tipifarnib-treated cells, respectively. Using phosphokinome profiling and immunofluorescence in the TF-1a cell line, we demonstrate that the drug combination is characterised by the activation of a DNA damage response (induction of ƎĀ³H2A.X and thr68 phosphorylation of chk2). Higher induction of ƎĀ³H2AX was found in CD34+CD38- than in CD34+CD38+ patient cells. In a model system, we show that dormancy impairs damage resolution, allowing accumulation of ƎĀ³H2AX foci. CONCLUSIONS: The chemosensitivity of the CD34+CD38- subset, combined with enhanced damage indicators, suggest that this subset is primed to favour programmed cell death as opposed to repairing damage. This interaction between tipifarnib and GO suggests a potential role in the treatment of AML.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Aminoglycosides/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD34/metabolism , Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/metabolism , Quinolones/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Gemtuzumab , Histones/metabolism , Humans , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Sialic Acid Binding Ig-like Lectin 3/metabolism , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/metabolism
13.
Indian J Clin Biochem ; 27(2): 121-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-23543587

ABSTRACT

The purpose of this study is to find out the frequency of TP53 mutations in acute myeloid leukemia (AML) patients and correlate sensitivity of drug response with TP53 mutations. In AML more than 90Ā % of cases comprise of wild type TP53. 94.2Ā % of TP53 mutations are found within exon 5-8 of which 73Ā % are point mutations. TP53 mutations were analysed with high resolution melting curve analysis. We analysed 106 AML samples of which we found nine mutations which represents 8.5Ā % mutation rate and found one rare SNP. The effect of TP53 mutations were studied on the chemosensitivity of two new drugs AZD115 and RHPS4, an Aurora Kinase B inhibitor and Telomerase inhibitor respectively. Four mutations were found out of 17 for RHPS4 stating significant (pĀ =Ā 0.002) increase in sensitivity and no mutation found in AZD1152 database, but need more study to get definite conclusion.

14.
Front Oncol ; 12: 840046, 2022.
Article in English | MEDLINE | ID: mdl-35707351

ABSTRACT

The protein kinase C (PKC) family of serine/threonine kinases are pleiotropic signaling regulators and are implicated in hematopoietic signaling and development. Only one isoform however, PKCƏĀµ, has oncogenic properties in solid cancers where it is associated with poor outcomes. Here we show that PKCƏĀµ protein is significantly overexpressed in acute myeloid leukemia (AML; 37% of patients). In addition, PKCƏĀµ expression in AML was associated with a significant reduction in complete remission induction and disease-free survival. Examination of the functional consequences of PKCƏĀµ overexpression in normal human hematopoiesis, showed that PKCƏĀµ promotes myeloid differentiation, particularly of the monocytic lineage, and decreased colony formation, suggesting that PKCƏĀµ does not act as an oncogene in hematopoietic cells. Rather, in AML cell lines, PKCƏĀµ overexpression selectively conferred resistance to the chemotherapeutic agent, daunorubicin, by reducing intracellular concentrations of this agent. Mechanistic analysis showed that PKCƏĀµ promoted the expression of the efflux pump, P-GP (ABCB1), and that drug efflux mediated by this transporter fully accounted for the daunorubicin resistance associated with PKCƏĀµ overexpression. Analysis of AML patient samples also showed a link between PKCƏĀµ and P-GP protein expression suggesting that PKCƏĀµ expression drives treatment resistance in AML by upregulating P-GP expression.

15.
Methods Protoc ; 5(2)2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35314662

ABSTRACT

Since mid-2020 there have been complexities and difficulties in the standardisation and administration of nasopharyngeal swabs. Coupled with the variable and/or poor accuracy of lateral flow devices, this has led to increased societal 'testing fatigue' and reduced confidence in test results. Consequently, asymptomatic individuals have developed reluctance towards repeat testing, which remains the best way to monitor COVID-19 cases in the wider population. On the other hand, saliva-based PCR, a non-invasive, highly sensitive, and accurate test suitable for everyone, is gaining momentum as a straightforward and reliable means of detecting SARS-CoV-2 in symptomatic and asymptomatic individuals. Here, we provide an itemised list of the equipment and reagents involved in the process of sample submission, inactivation and analysis, as well as a detailed description of how each of these steps is performed.

16.
Sci Rep ; 12(1): 11553, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35798820

ABSTRACT

Polymerase chain reaction (PCR) has proven to be the gold-standard for SARS-CoV-2 detection in clinical settings. The most common approaches rely on nasopharyngeal specimens obtained from swabs, followed by RNA extraction, reverse transcription and quantitative PCR. Although swab-based PCR is sensitive, swabbing is invasive and unpleasant to administer, reducing patient compliance for regular testing and resulting in an increased risk of improper sampling. To overcome these obstacles, we developed a non-invasive one-step RT-qPCR assay performed directly on saliva specimens. The University of Nottingham Asymptomatic Testing Service protocol simplifies sample collection and bypasses the need for RNA extraction, or additives, thus helping to encourage more regular testing and reducing processing time and costs. We have evaluated the assay against the performance criteria specified by the UK regulatory bodies and attained accreditation (BS EN ISO/IEC 17,025:2017) for SARS-CoV-2 diagnostic testing by the United Kingdom Accreditation Service. We observed a sensitivity of 1 viral copy per microlitre of saliva, and demonstrated a concordance of > 99.4% between our results and those of other accredited testing facilities. We concluded that saliva is a stable medium that allows for a highly precise, repeatable, and robust testing method.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/diagnosis , COVID-19 Testing , Clinical Laboratory Techniques/methods , Humans , Nasopharynx , RNA, Viral/analysis , RNA, Viral/genetics , SARS-CoV-2/genetics , Saliva/chemistry , Sensitivity and Specificity , Specimen Handling/methods
17.
BMC Cancer ; 11: 254, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21679421

ABSTRACT

BACKGROUND: Aurora kinases play an essential role in orchestrating chromosome alignment, segregation and cytokinesis during mitotic progression, with both aurora-A and B frequently over-expressed in a variety of human malignancies. Over-expression of the ABC drug transporter proteins P-glycoprotein (Pgp) and Breast cancer resistance protein (BCRP) is a major obstacle for chemotherapy in many tumour types with Pgp conferring particularly poor prognosis in acute myeloid leukaemia (AML). Barasertib-hQPA is a highly selective inhibitor of aurora-B kinase that has shown tumouricidal activity against a range tumour cell lines including those of leukaemic AML origin. METHODS: Effect of barasertib-hQPA on the pHH3 biomarker and cell viability was measured in a panel of leukaemic cell lines and 37 primary AML samples by flow cytometry. Pgp status was determined by flow cytometry and BCRP status by flow cytometry and real-time PCR. RESULTS: In this study we report the creation of the cell line OCI-AML3DNR, which over-expresses Pgp but not BCRP or multidrug resistance-associated protein (MRP), through prolonged treatment of OCI-AML3 cells with daunorubicin. We demonstrate that Pgp (OCI-AML3DNR and KG-1a) and BCRP (OCI-AML6.2) expressing AML cell lines are less sensitive to barasertib-hQPA induced pHH3 inhibition and subsequent loss of viability compared to transporter negative cell lines. We also show that barasertib-hQPA resistance in these cell lines can be reversed using known Pgp and BCRP inhibitors. We report that barasertib-hQPA is not an inhibitor of Pgp or BCRP, but by using 14[C]-barasertib-hQPA that it is effluxed by these transporters. Using phosphoHistone H3 (pHH3) as a biomarker of barasertib-hQPA responsiveness in primary AML blasts we determined that Pgp and BCRP positive primary samples were less sensitive to barasertib-hQPA induced pHH3 inhibition (p = <0.001) than samples without these transporters. However, we demonstrate that IC50 inhibition of pHH3 by barasertib-hQPA was achieved in 94.6% of these samples after 1 hour drug treatment, in contrast to the resistance of the cell lines. CONCLUSION: We conclude that Pgp and BCRP status and pHH3 down-regulation in patients treated with barasertib should be monitored in order to establish whether transporter-mediated efflux is sufficient to adversely impact on the efficacy of the agent.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , ATP-Binding Cassette Transporters/analysis , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/pathology , Neoplasm Proteins/antagonists & inhibitors , Organophosphates/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Daunorubicin/pharmacology , Female , Histones/metabolism , Humans , Inhibitory Concentration 50 , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Male , Neoplasm Proteins/analysis , Neoplasm Proteins/physiology , Organophosphates/pharmacokinetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/physiology , Quinazolines/pharmacokinetics
18.
Leuk Lymphoma ; 62(2): 281-290, 2021 02.
Article in English | MEDLINE | ID: mdl-33021411

ABSTRACT

Mutations in the highly similar genes B-cell translocation gene 1 (BTG1) and BTG2 are identified in approximately 10-15% of non-Hodgkin lymphoma cases, which may suggest a direct involvement of BTG1 and BTG2 in malignant transformation. However, it is unclear whether or how disease-associated mutations impair the function of these genes. Therefore, we selected 16 BTG1 variants based on in silico analysis. We then evaluated (i) the ability of these variants to interact with the known protein-binding partners CNOT7 and CNOT8, which encode the Caf1 catalytic subunit of the Ccr4-Not deadenylase complex; (ii) the activity of the variant proteins in cell cycle progression; (iii) translational repression; and (iv) mRNA degradation. Based on these analyses, we conclude that mutations in BTG1 may contribute to malignant transformation and tumor cell proliferation by interfering with its anti-proliferative activity and ability to interact with CNOT7 and CNOT8.


Subject(s)
Immediate-Early Proteins , Lymphoma, Non-Hodgkin , Cell Proliferation , Exoribonucleases , Humans , Lymphoma, Non-Hodgkin/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Binding , Receptors, CCR4 , Repressor Proteins , Transcription Factors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
19.
EJHaem ; 2(1): 40-47, 2021 Feb.
Article in English | MEDLINE | ID: mdl-35846088

ABSTRACT

Recent advances in treatment options for multiple myeloma (MM) have positive impact on patient survival. However, there is a short fall of rapid and reliable assays that can predict patient response to novel agents. The anti-apoptotic proteins B-cell lymphoma-2 (BCL-2) and myeloid cell leukaemia-1 (MCL-1), are necessary for MM survival, although most myelomas are more dependent on MCL-1. BCL-2 inhibition alone yields significant cytotoxicity in only a minority of cases, therefore targeting both proteins simultaneously, is a therapeutic option. Venetoclax and S63845 are BCL-2 and MCL-1 targeting BH3-mimetics which have demonstrated apoptotic synergy in MM. We investigated whether a novel short-term flow cytometric cytochrome c release assay could predict response to dual BH3-mimetic targeting in MM cells. Six human myeloma cell lines (HMCL) and seven primary samples were treated with venetoclax and S63845 alone or in combination. The 4-hour assay confirmed the drug combination was synergistic in all HMCL tested. Annexin-V data at 48 hours corresponded with 4-hour response verifying the assay as a predictor of drug sensitivity. All primary samples responded to the drug combination, including samples with 1q gain and t(4;14) translocation. Normal stem cells were unaffected by the drug combination. We have developed a novel assay with the potential to predict response to therapy in MM cells.

20.
J Med Microbiol ; 70(3)2021 Mar.
Article in English | MEDLINE | ID: mdl-33734960

ABSTRACT

Introduction. The COVID-19 pandemic, which began in 2020 is testing economic resilience and surge capacity of healthcare providers worldwide. At the time of writing, positive detection of the SARS-CoV-2 virus remains the only method for diagnosing COVID-19 infection. Rapid upscaling of national SARS-CoV-2 genome testing presented challenges: (1) Unpredictable supply chains of reagents and kits for virus inactivation, RNA extraction and PCR-detection of viral genomes. (2) Rapid time to result of <24 h is required in order to facilitate timely infection control measures.Hypothesis. Extraction-free sample processing would impact commercially available SARS-CoV-2 genome detection methods.Aim. We evaluated whether alternative commercially available kits provided sensitivity and accuracy of SARS-CoV-2 genome detection comparable to those used by regional National Healthcare Services (NHS).Methodology. We tested several detection methods and tested whether detection was altered by heat inactivation, an approach for rapid one-step viral inactivation and RNA extraction without chemicals or kits.Results. Using purified RNA, we found the CerTest VIASURE kit to be comparable to the Altona RealStar system currently in use, and further showed that both diagnostic kits performed similarly in the BioRad CFX96 and Roche LightCycler 480 II machines. Additionally, both kits were comparable to a third alternative using a combination of Quantabio qScript one-step Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR) mix and Centre for Disease Control and Prevention (CDC)-accredited N1 and N2 primer/probes when looking specifically at borderline samples. Importantly, when using the kits in an extraction-free protocol, following heat inactivation, we saw differing results, with the combined Quantabio-CDC assay showing superior accuracy and sensitivity. In particular, detection using the CDC N2 probe following the extraction-free protocol was highly correlated to results generated with the same probe following RNA extraction and reported clinically (n=127; R2=0.9259).Conclusion. Our results demonstrate that sample treatment can greatly affect the downstream performance of SARS-CoV-2 diagnostic kits, with varying impact depending on the kit. We also showed that one-step heat-inactivation methods could reduce time from swab receipt to outcome of test result. Combined, these findings present alternatives to the protocols in use and can serve to alleviate any arising supply-chain issues at different points in the workflow, whilst accelerating testing, and reducing cost and environmental impact.


Subject(s)
COVID-19 Nucleic Acid Testing , COVID-19/diagnosis , SARS-CoV-2/isolation & purification , Specimen Handling/methods , Culture Media , Hot Temperature , Humans , RNA, Viral/genetics , RNA, Viral/isolation & purification , Reagent Kits, Diagnostic , SARS-CoV-2/genetics , Sensitivity and Specificity , Virus Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL