Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38645166

ABSTRACT

Islet transplantation for treatment of diabetes is limited by availability of donor islets and requirements for immunosuppression. Stem cell-derived islets might circumvent these issues. SC-islets effectively control glucose metabolism post transplantation, but do not yet achieve full function in vitro with current published differentiation protocols. We aimed to identify markers of mature subpopulations of SC-ß cells by studying transcriptional changes associated with in vivo maturation of SC-ß cells using RNA-seq and co-expression network analysis. The ß cell-specific hormone islet amyloid polypeptide (IAPP) emerged as the top candidate to be such a marker. IAPP+ cells had more mature ß cell gene expression and higher cellular insulin content than IAPP- cells in vitro. IAPP+ INS+ cells were more stable in long-term culture than IAPP- INS+ cells and retained insulin expression after transplantation into mice. Finally, we conducted a small molecule screen to identify compounds that enhance IAPP expression. Aconitine up-regulated IAPP and could help to optimize differentiation protocols.

2.
Breast Cancer Res ; 22(1): 126, 2020 11 16.
Article in English | MEDLINE | ID: mdl-33198803

ABSTRACT

BACKGROUND: Activating transcription factor-2 (ATF2), a member of the leucine zipper family of DNA binding proteins, has been implicated as a tumour suppressor in breast cancer. However, its exact role in breast cancer endocrine resistance is still unclear. We have previously shown that silencing of ATF2 leads to a loss in the growth-inhibitory effects of tamoxifen in the oestrogen receptor (ER)-positive, tamoxifen-sensitive MCF7 cell line and highlighted that this multi-faceted transcription factor is key to the effects of tamoxifen in an endocrine sensitive model. In this work, we explored further the in vitro role of ATF2 in defining the resistance to endocrine treatment. MATERIALS AND METHODS: We knocked down ATF2 in TAMR, LCC2 and LCC9 tamoxifen-resistant breast cancer cell lines as well as the parental tamoxifen sensitive MCF7 cell line and investigated the effects on growth, colony formation and cell migration. We also performed a microarray gene expression profiling (Illumina Human HT12_v4) to explore alterations in gene expression between MCF7 and TAMRs after ATF2 silencing and confirmed gene expression changes by quantitative RT-PCR. RESULTS: By silencing ATF2, we observed a significant growth reduction of TAMR, LCC2 and LCC9 with no such effect observed with the parental MCF7 cells. ATF2 silencing was also associated with a significant inhibition of TAMR, LCC2 and LCC9 cell migration and colony formation. Interestingly, knockdown of ATF2 enhanced the levels of ER and ER-regulated genes, TFF1, GREB1, NCOA3 and PGR, in TAMR cells both at RNA and protein levels. Microarray gene expression identified a number of genes known to mediate tamoxifen resistance, to be differentially regulated by ATF2 in TAMR in relation to the parental MCF7 cells. Moreover, differential pathway analysis confirmed enhanced ER activity after ATF2 knockdown in TAMR cells. CONCLUSION: These data demonstrate that ATF2 silencing may overcome endocrine resistance and highlights further the dual role of this transcription factor that can mediate endocrine sensitivity and resistance by modulating ER expression and activity.


Subject(s)
Activating Transcription Factor 2/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Activating Transcription Factor 2/genetics , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , MCF-7 Cells , Oligonucleotide Array Sequence Analysis , Receptors, Estrogen/analysis , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
3.
Sci Rep ; 10(1): 546, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953501

ABSTRACT

Cytosine hydroxymethylation (5hmC) in mammalian DNA is the product of oxidation of methylated cytosines (5mC) by Ten-Eleven-Translocation (TET) enzymes. While it has been shown that the TETs influence 5mC metabolism, pluripotency and differentiation during early embryonic development, the functional relationship between gene expression and 5hmC in adult (somatic) stem cell differentiation is still unknown. Here we report that 5hmC levels undergo highly dynamic changes during adult stem cell differentiation from intestinal progenitors to differentiated intestinal epithelium. We profiled 5hmC and gene activity in purified mouse intestinal progenitors and differentiated progeny to identify 43425 differentially hydroxymethylated regions and 5325 differentially expressed genes. These differentially marked regions showed both losses and gains of 5hmC after differentiation, despite lower global levels of 5hmC in progenitor cells. In progenitors, 5hmC did not correlate with gene transcript levels, however, upon differentiation the global increase in 5hmC content showed an overall positive correlation with gene expression level as well as prominent associations with histone modifications that typify active genes and enhancer elements. Our data support a gene regulatory role for 5hmC that is predominant over its role in controlling DNA methylation states.


Subject(s)
5-Methylcytosine/analogs & derivatives , Cell Differentiation/drug effects , Cell Differentiation/genetics , Intestines/cytology , 5-Methylcytosine/pharmacology , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Animals , Mice
4.
Int J Cancer ; 145(12): 3389-3401, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31050820

ABSTRACT

Cancers occurring at the gastroesophageal junction (GEJ) are classified as predominantly esophageal or gastric, which is often difficult to decipher. We hypothesized that the transcriptomic profile might reveal molecular subgroups which could help to define the tumor origin and behavior beyond anatomical location. The gene expression profiles of 107 treatment-naïve, intestinal type, gastroesophageal adenocarcinomas were assessed by the Illumina-HTv4.0 beadchip. Differential gene expression (limma), unsupervised subgroup assignment (mclust) and pathway analysis (gage) were undertaken in R statistical computing and results were related to demographic and clinical parameters. Unsupervised assignment of the gene expression profiles revealed three distinct molecular subgroups, which were not associated with anatomical location, tumor stage or grade (p > 0.05). Group 1 was enriched for pathways involved in cell turnover, Group 2 was enriched for metabolic processes and Group 3 for immune-response pathways. Patients in group 1 showed the worst overall survival (p = 0.019). Key genes for the three subtypes were confirmed by immunohistochemistry. The newly defined intrinsic subtypes were analyzed in four independent datasets of gastric and esophageal adenocarcinomas with transcriptomic data available (RNAseq data: OCCAMS cohort, n = 158; gene expression arrays: Belfast, n = 63; Singapore, n = 191; Asian Cancer Research Group, n = 300). The subgroups were represented in the independent cohorts and pooled analysis confirmed the prognostic effect of the new subtypes. In conclusion, adenocarcinomas at the GEJ comprise three distinct molecular phenotypes which do not reflect anatomical location but rather inform our understanding of the key pathways expressed.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcriptome/genetics , Gene Expression Profiling/methods , Humans , Immunohistochemistry/methods , Phenotype , Prognosis , Prospective Studies
5.
Oncogene ; 38(7): 1136-1150, 2019 02.
Article in English | MEDLINE | ID: mdl-30237440

ABSTRACT

Elucidation of mechanisms underlying the increased androgen receptor (AR) activity and subsequent development of aggressive prostate cancer (PrCa) is pivotal in developing new therapies. Using a systems biology approach, we interrogated the AR-regulated proteome and identified PDZ binding kinase (PBK) as a novel AR-regulated protein that regulates full-length AR and AR variants (ARVs) activity in PrCa. PBK overexpression in aggressive PrCa is associated with early biochemical relapse and poor clinical outcome. In addition to its carboxy terminus ligand-binding domain, PBK directly interacts with the amino terminus transactivation domain of the AR to stabilise it thereby leading to increased AR protein expression observed in PrCa. Transcriptome sequencing revealed that PBK is a mediator of global AR signalling with key roles in regulating tumour invasion and metastasis. PBK inhibition decreased growth of PrCa cell lines and clinical specimen cultured ex vivo. We uncovered a novel interplay between AR and PBK that results in increased AR and ARVs expression that executes AR-mediated growth and progression of PrCa, with implications for the development of PBK inhibitors for the treatment of aggressive PrCa.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Cell Line, Tumor , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Male , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptors, Androgen/genetics
6.
Mol Genet Metab Rep ; 15: 124-126, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30023302

ABSTRACT

Mucopolysaccharidoses (MPS) type III also termed as Sanfillipo syndrome, involves defect in enzymes required for degradation of heparan sulphate. We report a clinical case of MPS-III later followed by genetic investigation for MPS-III genes SGSH, NAGLU, HGSNAT and GNS. It allowed us to identify a novel and likely pathogenic variant p. G205R in SGSH. Protein based Inslico prediction and protein modelling suggests aberration of helical structure of SGSH protein and reduced binding affinity for its substrate.

7.
Mol Metab ; 10: 74-86, 2018 04.
Article in English | MEDLINE | ID: mdl-29472102

ABSTRACT

OBJECTIVE: Dedifferentiation could explain reduced functional pancreatic ß-cell mass in type 2 diabetes (T2D). METHODS: Here we model human ß-cell dedifferentiation using growth factor stimulation in the human ß-cell line, EndoC-ßH1, and human pancreatic islets. RESULTS: Fibroblast growth factor 2 (FGF2) treatment reduced expression of ß-cell markers, (INS, MAFB, SLC2A2, SLC30A8, and GCK) and activated ectopic expression of MYC, HES1, SOX9, and NEUROG3. FGF2-induced dedifferentiation was time- and dose-dependent and reversible upon wash-out. Furthermore, FGF2 treatment induced expression of TNFRSF11B, a decoy receptor for RANKL and protected ß-cells against RANKL signaling. Finally, analyses of transcriptomic data revealed increased FGF2 expression in ductal, endothelial, and stellate cells in pancreas from T2D patients, whereas FGFR1, SOX,9 and HES1 expression increased in islets from T2D patients. CONCLUSIONS: We thus developed an FGF2-induced model of human ß-cell dedifferentiation, identified new markers of dedifferentiation, and found evidence for increased pancreatic FGF2, FGFR1, and ß-cell dedifferentiation in T2D.


Subject(s)
Cell Dedifferentiation , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/cytology , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Fibroblast Growth Factor 2/pharmacology , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
8.
Eur J Med Chem ; 134: 218-229, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28415011

ABSTRACT

Adenosine induces bronchial hyperresponsiveness and inflammation in asthmatics through activation of A2B adenosine receptor (A2BAdoR). Selective antagonists have been shown to attenuate airway reactivity and improve inflammatory conditions in pre-clinical studies. Hence, the identification of novel, potent and selective A2BAdoR antagonist may be beneficial for the potential treatment of asthma and Chronic Obstructive Pulmonary Disease (COPD). Towards this effort, we explored several prop-2-ynylated C8-aryl or heteroaryl substitutions on xanthine chemotype and found that 1-prop-2-ynyl-1H-pyrazol-4-yl moiety was better tolerated at the C8 position. Compound 59, exhibited binding affinity (Ki) of 62 nM but was non-selective for A2BAdoR over other AdoRs. Incorporation of substituted phenyl on the terminal acetylene increased the binding affinity (Ki) significantly to <10 nM. Various substitutions on terminal phenyl group and different alkyl substitutions on N-1 and N-3 were explored to improve the potency, selectivity for A2BAdoR and the solubility. In general, compounds with meta-substituted phenyl provided better selectivity for A2BAdoR compared to that of para-substituted analogs. Substitutions such as basic amines like pyrrolidine, piperidine, piperazine or cycloalkyls with polar group were tried on terminal acetylene, keeping in mind the poor solubility of xanthine analogs in general. However, these substitutions led to a decrease in affinity compared to compound 59. Subsequent SAR optimization resulted in identification of compound 46 with high human A2BAdoR affinity (Ki = 13 nM), selectivity against other AdoR subtypes and with good pharmacokinetic properties. It was found to be a potent functional A2BAdoR antagonist with a Ki of 8 nM in cAMP assay in hA2B-HEK293 cells and an IC50 of 107 nM in IL6 assay in NIH-3T3 cells. Docking study was performed to rationalize the observed affinity data. Structure-activity relationship (SAR) studies also led to identification of compound 36 as a potent A2BAdoR antagonist with Ki of 1.8 nM in cAMP assay and good aqueous solubility of 529 µM at neutral pH. Compound 46 was further tested for in vivo efficacy and found to be efficacious in ovalbumin-induced allergic asthma model in mice.


Subject(s)
Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/therapeutic use , Asthma/drug therapy , Receptor, Adenosine A2B/metabolism , Xanthine/chemistry , Xanthine/therapeutic use , Adenosine A2 Receptor Antagonists/metabolism , Adenosine A2 Receptor Antagonists/pharmacokinetics , Animals , Asthma/chemically induced , Asthma/metabolism , Dogs , Drug Design , Hep G2 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Docking Simulation , Ovalbumin , Rats , Receptor, Adenosine A2B/chemistry , Xanthine/metabolism , Xanthine/pharmacokinetics
9.
Bioorg Med Chem ; 25(6): 1963-1975, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28238512

ABSTRACT

Multipronged approach was used to synthesize a library of diverse C-8 cyclopentyl hypoxanthine analogs from a common intermediate III. Several potent and selective compounds were identified and evaluated for pharmacokinetic (PK) properties in Wistar rats. One of the compounds 14 with acceptable PK parameters was selected for testing in in vivo primary acute diuresis model. The compound demonstrated significant diuretic activity in this model.


Subject(s)
Adenosine A1 Receptor Antagonists/chemistry , Adenosine A1 Receptor Antagonists/pharmacology , Hypoxanthines/chemistry , Hypoxanthines/pharmacology , Adenosine A1 Receptor Antagonists/chemical synthesis , Adenosine A1 Receptor Antagonists/pharmacokinetics , Animals , Carbon-13 Magnetic Resonance Spectroscopy , Chromatography, Liquid , Drug Design , HEK293 Cells , Humans , Hypoxanthines/chemical synthesis , Hypoxanthines/pharmacokinetics , Male , Mass Spectrometry , Proton Magnetic Resonance Spectroscopy , Radioligand Assay , Rats , Rats, Wistar
10.
J Med Chem ; 60(2): 681-694, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28055204

ABSTRACT

Our initial structure-activity relationship studies on 7-methoxy-4-morpholino-benzothiazole derivatives featured by aryloxy-2-methylpropanamide moieties at the 2-position led to identification of compound 25 as a potent and selective A2A adenosine receptor (A2AAdoR) antagonist with reasonable ADME and pharmacokinetic properties. However, poor intrinsic solubility and low to moderate oral bioavailability made this series unsuitable for further development. Further optimization using structure-based drug design approach resulted in discovery of potent and selective adenosine A2A receptor antagonists bearing substituted 1-methylcyclohexyl-carboxamide groups at position 2 of the benzothiazole scaffold and endowed with better solubility and oral bioavailability. Compounds 41 and 49 demonstrated a number of positive attributes with respect to in vitro ADME properties. Both compounds displayed good pharmacokinetic properties with 63% and 61% oral bioavailability, respectively, in rat. Further, compound 49 displayed oral efficacy in 6-OHDA lesioned rat model of Parkinson diseases.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Benzothiazoles/pharmacology , Cyclohexanols/pharmacology , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/pharmacokinetics , Administration, Oral , Animals , Antiparkinson Agents/chemical synthesis , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/pharmacology , Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacokinetics , Cyclohexanols/chemical synthesis , Cyclohexanols/pharmacokinetics , Drug Design , HEK293 Cells , Humans , Levodopa/pharmacology , Male , Microsomes, Liver/metabolism , Molecular Docking Simulation , Rats, Wistar , Structure-Activity Relationship
11.
Eur J Med Chem ; 127: 986-996, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27842891

ABSTRACT

A2BAdoR is a low affinity adenosine receptor that functions by Gs mediated elevation of cAMP and subsequent downstream signaling. The receptor has been implicated in lung inflammatory disorders like COPD and asthma. Several potent and selective A2BAdoR antagonists have been reported in literature, however most of the compounds suffer from poor pharmacokinetic profile. Therefore, with the aim to identify novel, potent and selective A2BAdoR antagonists with improved pharmacokinetic properties, we first explored more constrained form of MRS-1754 (4). To improve the metabolic stability, several linker modifications were attempted as replacement of amide linker along with different phenyl or other heteroaryls between C8 position of xanthine head group and terminal phenyl ring. SAR optimization resulted in identification of two novel A2BAdoR antagonists, 8-{1-[5-Oxo-1-(4-trifluoromethyl-phenyl)-pyrrolidin-3-ylmethyl]-1H-pyrazol-4-yl}-1,3-dipropyl-xanthine (31) and 8-(1-{2-Oxo-2-[4-(3-trifluoromethyl-phenyl)-piperazin-1-yl]-ethyl}-1H-pyrazol-4-yl)-1,3-dipropyl-xanthine (65), with high binding affinity (Ki = 1 and 1.5 nM, respectively) and selectivity for A2BAdoR with very good functional potency of 0.9 nM and 4 nM, respectively. Compound 31 and 65 also displayed good pharmacokinetic properties in mice with 27% and 65% oral bioavailability respectively. When evaluated in in vivo mice model of asthma, compound 65 also inhibited airway inflammation and airway reactivity in ovalbumin induced allergic asthma at 3 mpk dose.


Subject(s)
Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/pharmacology , Drug Design , Receptor, Adenosine A2B/metabolism , Xanthine/chemical synthesis , Xanthine/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Animals , Brain/drug effects , Brain/metabolism , Chemistry Techniques, Synthetic , Male , Mice , Structure-Activity Relationship , Xanthine/chemistry
12.
Endocr Relat Cancer ; 23(10): 797-812, 2016 10.
Article in English | MEDLINE | ID: mdl-27578825

ABSTRACT

Due to increased sensitivity, the expression of circulating nucleotides is rapidly gaining popularity in cancer diagnosis. Whole blood mRNA has been used in studies on a number of cancers, most notably two separate studies that used whole blood mRNA to define non-overlapping signatures of prostate cancer that has become castration independent. Prostate cancer is known to rely on androgens for initial growth, and there is increasing evidence on the importance of the androgen axis in advanced disease. Using whole blood mRNA samples from patients with prostate cancer, we have identified the four-gene panel of FAM129A, MME, KRT7 and SOD2 in circulating mRNA that are differentially expressed in a discovery cohort of metastatic samples. Validation of these genes at the mRNA and protein level was undertaken in additional cohorts defined by risk of relapse following surgery and hormone status. All the four genes were downregulated at the mRNA level in the circulation and in primary tissue, but this was not always reflected in tissue protein expression. MME demonstrated significant differences in the hormone cohorts, whereas FAM129A is downregulated at the mRNA level but is raised at the protein level in tumours. Using published ChIP-seq data, we have demonstrated that this may be due to AR binding at the FAM129A and MME loci in multiple cell lines. These data suggest that whole blood mRNA of androgen-regulated genes has the potential to be used for diagnosis and monitoring of prostate cancer.


Subject(s)
Androgens/pharmacology , Prostatic Neoplasms/genetics , RNA, Messenger/blood , Transcriptome/drug effects , Aged , Aged, 80 and over , Blood Chemical Analysis/methods , Case-Control Studies , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Microarray Analysis , Middle Aged , Prostatic Neoplasms/blood , RNA, Messenger/analysis
13.
Nat Cell Biol ; 18(9): 979-92, 2016 09.
Article in English | MEDLINE | ID: mdl-27525720

ABSTRACT

Senescence, a persistent form of cell-cycle arrest, is often associated with a diverse secretome, which provides complex functionality for senescent cells within the tissue microenvironment. We show that oncogene-induced senescence is accompanied by a dynamic fluctuation of NOTCH1 activity, which drives a TGF-ß-rich secretome, while suppressing the senescence-associated pro-inflammatory secretome through inhibition of C/EBPß. NOTCH1 and NOTCH1-driven TGF-ß contribute to 'lateral induction of senescence' through a juxtacrine NOTCH-JAG1 pathway. In addition, NOTCH1 inhibition during senescence facilitates upregulation of pro-inflammatory cytokines, promoting lymphocyte recruitment and senescence surveillance in vivo. As enforced activation of NOTCH1 signalling confers a near mutually exclusive secretory profile compared with typical senescence, our data collectively indicate that the dynamic alteration of NOTCH1 activity during senescence dictates a functional balance between these two distinct secretomes: one representing TGF-ß and the other pro-inflammatory cytokines, highlighting that NOTCH1 is a temporospatial controller of secretome composition.


Subject(s)
Cell Cycle Checkpoints/physiology , Receptor, Notch1/metabolism , Animals , Cell Line, Tumor , Cellular Senescence , Humans , Mice, Transgenic , Receptor, Notch1/genetics , Transforming Growth Factor beta/metabolism
14.
Nat Commun ; 7: 10406, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26832224

ABSTRACT

Long noncoding RNAs (lncRNAs) regulate gene expression via their RNA product or through transcriptional interference, yet a strategy to differentiate these two processes is lacking. To address this, we used multiple small interfering RNAs (siRNAs) to silence GNG12-AS1, a nuclear lncRNA transcribed in an antisense orientation to the tumour-suppressor DIRAS3. Here we show that while most siRNAs silence GNG12-AS1 post-transcriptionally, siRNA complementary to exon 1 of GNG12-AS1 suppresses its transcription by recruiting Argonaute 2 and inhibiting RNA polymerase II binding. Transcriptional, but not post-transcriptional, silencing of GNG12-AS1 causes concomitant upregulation of DIRAS3, indicating a function in transcriptional interference. This change in DIRAS3 expression is sufficient to impair cell cycle progression. In addition, the reduction in GNG12-AS1 transcripts alters MET signalling and cell migration, but these are independent of DIRAS3. Thus, differential siRNA targeting of a lncRNA allows dissection of the functions related to the process and products of its transcription.


Subject(s)
GTP-Binding Protein gamma Subunits/metabolism , RNA, Long Noncoding/metabolism , rho GTP-Binding Proteins/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Cell Cycle , GTP-Binding Protein gamma Subunits/genetics , Humans , Oligonucleotide Array Sequence Analysis , Protein Isoforms , RNA Interference , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , RNA, Long Noncoding/genetics , rho GTP-Binding Proteins/genetics
15.
J Natl Cancer Inst ; 108(5)2016 May.
Article in English | MEDLINE | ID: mdl-26657335

ABSTRACT

BACKGROUND: The androgen receptor (AR) is a major drug target in prostate cancer (PCa). We profiled the AR-regulated kinome to identify clinically relevant and druggable effectors of AR signaling. METHODS: Using genome-wide approaches, we interrogated all AR regulated kinases. Among these, choline kinase alpha (CHKA) expression was evaluated in benign (n = 195), prostatic intraepithelial neoplasia (PIN) (n = 153) and prostate cancer (PCa) lesions (n = 359). We interrogated how CHKA regulates AR signaling using biochemical assays and investigated androgen regulation of CHKA expression in men with PCa, both untreated (n = 20) and treated with an androgen biosynthesis inhibitor degarelix (n = 27). We studied the effect of CHKA inhibition on the PCa transcriptome using RNA sequencing and tested the effect of CHKA inhibition on cell growth, clonogenic survival and invasion. Tumor xenografts (n = 6 per group) were generated in mice using genetically engineered prostate cancer cells with inducible CHKA knockdown. Data were analyzed with χ(2) tests, Cox regression analysis, and Kaplan-Meier methods. All statistical tests were two-sided. RESULTS: CHKA expression was shown to be androgen regulated in cell lines, xenografts, and human tissue (log fold change from 6.75 to 6.59, P = .002) and was positively associated with tumor stage. CHKA binds directly to the ligand-binding domain (LBD) of AR, enhancing its stability. As such, CHKA is the first kinase identified as an AR chaperone. Inhibition of CHKA repressed the AR transcriptional program including pathways enriched for regulation of protein folding, decreased AR protein levels, and inhibited the growth of PCa cell lines, human PCa explants, and tumor xenografts. CONCLUSIONS: CHKA can act as an AR chaperone, providing, to our knowledge, the first evidence for kinases as molecular chaperones, making CHKA both a marker of tumor progression and a potential therapeutic target for PCa.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Choline Kinase/metabolism , Molecular Chaperones , Molecular Targeted Therapy/methods , Prostatectomy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Receptors, Androgen/metabolism , Signal Transduction , Aged , Animals , Choline Kinase/antagonists & inhibitors , Choline Kinase/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Grading , Neoplasm Staging , Proportional Hazards Models , Prostatectomy/methods , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Sequence Analysis, DNA , Xenograft Model Antitumor Assays
16.
Metabolomics ; 11(6): 1848-1863, 2015.
Article in English | MEDLINE | ID: mdl-26491426

ABSTRACT

To investigate metabolic changes during cellular transformation, we used a 1H NMR based metabolite-metabolite correlation analysis (MMCA) method, which permits analysis of homeostatic mechanisms in cells at the steady state, in an inducible cell transformation model. Transcriptomic data were used to further explain the results. Transformed cells showed many more metabolite-metabolite correlations than control cells. Some had intuitively plausible explanations: a shift from glycolysis to amino acid oxidation after transformation was accompanied by a strongly positive correlation between glucose and glutamine and a strongly negative one between lactate and glutamate; there were also many correlations between the branched chain amino acids and the aromatic amino acids. Others remain puzzling: after transformation strong positive correlations developed between choline and a group of five amino acids, whereas the same amino acids showed negative correlations with phosphocholine, a membrane phospholipid precursor. MMCA in conjunction with transcriptome analysis has opened a new window into the metabolome.

18.
Nature ; 523(7560): 313-7, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26153859

ABSTRACT

Progesterone receptor (PR) expression is used as a biomarker of oestrogen receptor-α (ERα) function and breast cancer prognosis. Here we show that PR is not merely an ERα-induced gene target, but is also an ERα-associated protein that modulates its behaviour. In the presence of agonist ligands, PR associates with ERα to direct ERα chromatin binding events within breast cancer cells, resulting in a unique gene expression programme that is associated with good clinical outcome. Progesterone inhibited oestrogen-mediated growth of ERα(+) cell line xenografts and primary ERα(+) breast tumour explants, and had increased anti-proliferative effects when coupled with an ERα antagonist. Copy number loss of PGR, the gene coding for PR, is a common feature in ERα(+) breast cancers, explaining lower PR levels in a subset of cases. Our findings indicate that PR functions as a molecular rheostat to control ERα chromatin binding and transcriptional activity, which has important implications for prognosis and therapeutic interventions.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/drug effects , Chromatin/genetics , Chromatin/metabolism , DNA Copy Number Variations/genetics , Disease Progression , Estrogen Receptor alpha/antagonists & inhibitors , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Mice , Progesterone/metabolism , Progesterone/pharmacology , Protein Binding/drug effects , Receptors, Progesterone/genetics , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
19.
PLoS Genet ; 11(3): e1005053, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25790137

ABSTRACT

The downstream functions of the DNA binding tumor suppressor p53 vary depending on the cellular context, and persistent p53 activation has recently been implicated in tumor suppression and senescence. However, genome-wide information about p53-target gene regulation has been derived mostly from acute genotoxic conditions. Using ChIP-seq and expression data, we have found distinct p53 binding profiles between acutely activated (through DNA damage) and chronically activated (in senescent or pro-apoptotic conditions) p53. Compared to the classical 'acute' p53 binding profile, 'chronic' p53 peaks were closely associated with CpG-islands. Furthermore, the chronic CpG-island binding of p53 conferred distinct expression patterns between senescent and pro-apoptotic conditions. Using the p53 targets seen in the chronic conditions together with external high-throughput datasets, we have built p53 networks that revealed extensive self-regulatory 'p53 hubs' where p53 and many p53 targets can physically interact with each other. Integrating these results with public clinical datasets identified the cancer-associated lipogenic enzyme, SCD, which we found to be directly repressed by p53 through the CpG-island promoter, providing a mechanistic link between p53 and the 'lipogenic phenotype', a hallmark of cancer. Our data reveal distinct phenotype associations of chronic p53 targets that underlie specific gene regulatory mechanisms.


Subject(s)
DNA Methylation/genetics , DNA-Binding Proteins/genetics , Protein Interaction Maps/genetics , Tumor Suppressor Protein p53/genetics , Aging/genetics , Apoptosis/genetics , Cell Line , CpG Islands/genetics , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Phenotype , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Tumor Suppressor Protein p53/metabolism
20.
N Engl J Med ; 372(7): 601-612, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25671252

ABSTRACT

BACKGROUND: Cancers result from the accumulation of somatic mutations, and their properties are thought to reflect the sum of these mutations. However, little is known about the effect of the order in which mutations are acquired. METHODS: We determined mutation order in patients with myeloproliferative neoplasms by genotyping hematopoietic colonies or by means of next-generation sequencing. Stem cells and progenitor cells were isolated to study the effect of mutation order on mature and immature hematopoietic cells. RESULTS: The age at which a patient presented with a myeloproliferative neoplasm, acquisition of JAK2 V617F homozygosity, and the balance of immature progenitors were all influenced by mutation order. As compared with patients in whom the TET2 mutation was acquired first (hereafter referred to as "TET2-first patients"), patients in whom the Janus kinase 2 (JAK2) mutation was acquired first ("JAK2-first patients") had a greater likelihood of presenting with polycythemia vera than with essential thrombocythemia, an increased risk of thrombosis, and an increased sensitivity of JAK2-mutant progenitors to ruxolitinib in vitro. Mutation order influenced the proliferative response to JAK2 V617F and the capacity of double-mutant hematopoietic cells and progenitor cells to generate colony-forming cells. Moreover, the hematopoietic stem-and-progenitor-cell compartment was dominated by TET2 single-mutant cells in TET2-first patients but by JAK2-TET2 double-mutant cells in JAK2-first patients. Prior mutation of TET2 altered the transcriptional consequences of JAK2 V617F in a cell-intrinsic manner and prevented JAK2 V617F from up-regulating genes associated with proliferation. CONCLUSIONS: The order in which JAK2 and TET2 mutations were acquired influenced clinical features, the response to targeted therapy, the biology of stem and progenitor cells, and clonal evolution in patients with myeloproliferative neoplasms. (Funded by Leukemia and Lymphoma Research and others.).


Subject(s)
DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/physiology , Janus Kinase 2/genetics , Mutation , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins/genetics , Age of Onset , Cell Proliferation/genetics , DNA Mutational Analysis , Dioxygenases , Gene Expression , Homozygote , Humans , Polycythemia Vera/genetics , Thrombocythemia, Essential/genetics , Thrombosis/genetics , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...