Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
Add more filters

Publication year range
1.
Ann Oncol ; 30(7): 1071-1079, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31090900

ABSTRACT

BACKGROUND: Whole-genome sequencing (WGS) is a powerful method for revealing the diversity and complexity of the somatic mutation burden of tumours. Here, we investigated the utility of tumour and matched germline WGS for understanding aetiology and treatment opportunities for high-risk individuals with familial breast cancer. PATIENTS AND METHODS: We carried out WGS on 78 paired germline and tumour DNA samples from individuals carrying pathogenic variants in BRCA1 (n = 26) or BRCA2 (n = 22) or from non-carriers (non-BRCA1/2; n = 30). RESULTS: Matched germline/tumour WGS and somatic mutational signature analysis revealed patients with unreported, dual pathogenic germline variants in cancer risk genes (BRCA1/BRCA2; BRCA1/MUTYH). The strategy identified that 100% of tumours from BRCA1 carriers and 91% of tumours from BRCA2 carriers exhibited biallelic inactivation of the respective gene, together with somatic mutational signatures suggestive of a functional deficiency in homologous recombination. A set of non-BRCA1/2 tumours also had somatic signatures indicative of BRCA-deficiency, including tumours with BRCA1 promoter methylation, and tumours from carriers of a PALB2 pathogenic germline variant and a BRCA2 variant of uncertain significance. A subset of 13 non-BRCA1/2 tumours from early onset cases were BRCA-proficient, yet displayed complex clustered structural rearrangements associated with the amplification of oncogenes and pathogenic germline variants in TP53, ATM and CHEK2. CONCLUSIONS: Our study highlights the role that WGS of matched germline/tumour DNA and the somatic mutational signatures can play in the discovery of pathogenic germline variants and for providing supporting evidence for variant pathogenicity. WGS-derived signatures were more robust than germline status and other genomic predictors of homologous recombination deficiency, thus impacting the selection of platinum-based or PARP inhibitor therapy. In this first examination of non-BRCA1/2 tumours by WGS, we illustrate the considerable heterogeneity of these tumour genomes and highlight that complex genomic rearrangements may drive tumourigenesis in a subset of cases.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Germ-Line Mutation , Adult , Breast Neoplasms/pathology , DNA, Neoplasm/genetics , Fanconi Anemia Complementation Group N Protein/genetics , Female , Genetic Predisposition to Disease , Humans , Middle Aged , Prognosis , Whole Genome Sequencing/methods
3.
Cancer Metastasis Rev ; 31(1-2): 1-19, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22138778

ABSTRACT

Inherited mutations are known to cause familial cancers. However, the cause of sporadic cancers, which likely represent the majority of cancers, is yet to be elucidated. Sporadic cancers contain somatic mutations (including oncogenic mutations); however, the origin of these mutations is unclear. An intriguing possibility is that a stable alteration occurs in somatic cells prior to oncogenic mutations and promotes the subsequent accumulation of oncogenic mutations. This review explores the possible role of prions and protein-only inheritance in cancer. Genetic studies using lower eukaryotes, primarily yeast, have identified a large number of proteins as prions that confer dominant phenotypes with cytoplasmic (non-Mendelian) inheritance. Many of these have mammalian functional homologs. The human prion protein (PrP) is known to cause neurodegenerative diseases and has now been found to be upregulated in multiple cancers. PrP expression in cancer cells contributes to cancer progression and resistance to various cancer therapies. Epigenetic changes in the gene expression and hyperactivation of MAP kinase signaling, processes that in lower eukaryotes are affected by prions, play important roles in oncogenesis in humans. Prion phenomena in yeast appear to be influenced by stresses, and there is considerable evidence of the association of some amyloids with biologically positive functions. This suggests that if protein-only somatic inheritance exists in mammalian cells, it might contribute to cancer phenotypes. Here, we highlight evidence in the literature for an involvement of prion or prion-like mechanisms in cancer and how they may in the future be viewed as diagnostic markers and potential therapeutic targets.


Subject(s)
Neoplasms/genetics , Neoplasms/metabolism , Prions/metabolism , Heredity , Humans , Models, Biological , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasms/diagnosis , Neoplasms/therapy
4.
Nat Genet ; 27(3): 247-54, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11242102

ABSTRACT

To ensure the high-fidelity transmission of genetic information, cells have evolved mechanisms to monitor genome integrity. Cells respond to DNA damage by activating a complex DNA-damage-response pathway that includes cell-cycle arrest, the transcriptional and post-transcriptional activation of a subset of genes including those associated with DNA repair, and, under some circumstances, the triggering of programmed cell death. An inability to respond properly to, or to repair, DNA damage leads to genetic instability, which in turn may enhance the rate of cancer development. Indeed, it is becoming increasingly clear that deficiencies in DNA-damage signaling and repair pathways are fundamental to the etiology of most, if not all, human cancers. Here we describe recent progress in our understanding of how cells detect and signal the presence and repair of one particularly important form of DNA damage induced by ionizing radiation-the DNA double-strand break (DSB). Moreover, we discuss how tumor suppressor proteins such as p53, ATM, Brca1 and Brca2 have been linked to such pathways, and how accumulating evidence is connecting deficiencies in cellular responses to DNA DSBs with tumorigenesis.


Subject(s)
DNA Damage , DNA Repair , Neoplasms/genetics , Neoplasms/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein/metabolism , BRCA2 Protein , Cell Cycle , Cell Cycle Proteins , DNA Repair/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins , Humans , Neoplasm Proteins/metabolism , Neoplasms/etiology , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
5.
Nat Genet ; 25(1): 115-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10802669

ABSTRACT

Mutations in the gene ATM are responsible for the genetic disorder ataxia-telangiectasia (A-T), which is characterized by cerebellar dysfunction, radiosensitivity, chromosomal instability and cancer predisposition. Both the A-T phenotype and the similarity of the ATM protein to other DNA-damage sensors suggests a role for ATM in biochemical pathways involved in the recognition, signalling and repair of DNA double-strand breaks (DSBs). There are strong parallels between the pattern of radiosensitivity, chromosomal instability and cancer predisposition in A-T patients and that in patients with Nijmegen breakage syndrome (NBS). The protein defective in NBS, nibrin (encoded by NBS1), forms a complex with MRE11 and RAD50 (refs 1,2). This complex localizes to DSBs within 30 minutes after cellular exposure to ionizing radiation (IR) and is observed in brightly staining nuclear foci after a longer period of time. The overlap between clinical and cellular phenotypes in A-T and NBS suggests that ATM and nibrin may function in the same biochemical pathway. Here we demonstrate that nibrin is phosphorylated within one hour of treatment of cells with IR. This response is abrogated in A-T cells that either do not express ATM protein or express near full-length mutant protein. We also show that ATM physically interacts with and phosphorylates nibrin on serine 343 both in vivo and in vitro. Phosphorylation of this site appears to be functionally important because mutated nibrin (S343A) does not completely complement radiosensitivity in NBS cells. ATM phosphorylation of nibrin does not affect nibrin-MRE11-RAD50 association as revealed by radiation-induced foci formation. Our data provide a biochemical explanation for the similarity in phenotype between A-T and NBS.


Subject(s)
Cell Cycle Proteins/radiation effects , Gamma Rays , Nuclear Proteins , Protein Serine-Threonine Kinases/radiation effects , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line , Chromosome Breakage/genetics , DNA-Binding Proteins , Genetic Predisposition to Disease/genetics , Humans , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins
6.
Nat Genet ; 20(4): 398-400, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9843217

ABSTRACT

The human genetic disorder ataxia-telangiectasia (AT) is characterized by immunodeficiency, progressive cerebellar ataxia, radiosensitivity, cell cycle checkpoint defects and cancer predisposition. The gene mutated in this syndrome, ATM (for AT mutated), encodes a protein containing a phosphatidyl-inositol 3-kinase (PI-3 kinase)-like domain. ATM also contains a proline-rich region and a leucine zipper, both of which implicate this protein in signal transduction. The proline-rich region has been shown to bind to the SH3 domain of c-Abl, which facilitates its phosphorylation and activation by ATM. Previous results have demonstrated that AT cells are defective in the G1/S checkpoint activated after radiation damage and that this defect is attributable to a defective p53 signal transduction pathway. We report here direct interaction between ATM and p53 involving two regions in ATM, one at the amino terminus and the other at the carboxy terminus, corresponding to the PI-3 kinase domain. Recombinant ATM protein phosphorylates p53 on serine 15 near the N terminus. Furthermore, ectopic expression of ATM in AT cells restores normal ionizing radiation (IR)-induced phosphorylation of p53, whereas expression of ATM antisense RNA in control cells abrogates the rapid IR-induced phosphorylation of p53 on serine 15. These results demonstrate that ATM can bind p53 directly and is responsible for its serine 15 phosphorylation, thereby contributing to the activation and stabilization of p53 during the IR-induced DNA damage response.


Subject(s)
Protein Serine-Threonine Kinases , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ataxia Telangiectasia Mutated Proteins , Binding Sites , Cell Cycle Proteins , DNA-Binding Proteins , Humans , Phosphorylation , Protein Binding , Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins
7.
Mamm Genome ; 22(3-4): 235-48, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21347845

ABSTRACT

An N-ethyl-N-nitrosourea random mutation screen was used to identify recessive modifiers of gene silencing in the mouse using an epigenetically sensitive reporter transgene. One of the mutant lines, MommeR1, was identified as a suppressor of variegation and it showed female-specific age-associated infertility in homozygotes. Linkage analysis identified a region on chromosome 10, containing the Foxo3a gene, previously shown to play a critical role in female gametogenesis. Foxo3a is a transcription factor with roles in cell cycle control, apoptosis, neural and hematopoietic cell differentiation, and DNA repair. Sequencing of the Foxo3a gene in MommeR1 mice revealed a point mutation that causes an amino acid substitution in the highly conserved Forkhead DNA-binding domain. In vitro transcription assays showed that the point mutation causes loss of FOXO3a transactivation activity. Compound heterozygotes made with Foxo3a-null mice (carrying the targeted deletion of exon 2) displayed complementation with respect to both the activation of the reporter transgene and defects in folliculogenesis similar to those seen in MommeR1 homozygotes, supporting the conclusion that this is the causative mutation. Approximately one in six female MommeR1 homozygotes develop teratomas, a phenotype not reported in Foxo3a-null mice. Ovulated oocytes from MommeR1 homozygotes display a number of abnormalities. The MommeR1 mice provide a novel platform to investigate teratocarcinogenesis and link Foxo3a with parthenogenesis and ovarian cancer. The finding of Foxo3a as a modifier of epigenetic reprogramming is discussed.


Subject(s)
Forkhead Transcription Factors/genetics , Mutation, Missense , Oocytes/cytology , Ovarian Neoplasms/genetics , Teratoma/genetics , Amino Acid Sequence , Animals , Base Sequence , Disease Models, Animal , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Silencing , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Oocytes/metabolism , Ovarian Neoplasms/metabolism , Point Mutation , Teratoma/metabolism
8.
VideoGIE ; 6(12): 546-548, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34917866

ABSTRACT

Video 1Pursuit of a pancreatic mass: autoimmune pancreatitis mimicking pancreatic cancer. EUS features of autoimmune pancreatitis in an older man who presented with obstructive jaundice and pancreatic mass.

9.
J Exp Med ; 191(9): 1459-66, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10790421

ABSTRACT

Recurrent herpes simplex virus type 1 (HSV-1) disease usually results from reactivation of latent virus in sensory neurons and transmission to peripheral sites. Therefore, defining the mechanisms that maintain HSV-1 in a latent state in sensory neurons may provide new approaches to reducing susceptibility to recurrent herpetic disease. After primary HSV-1 corneal infection, CD8(+) T cells infiltrate the trigeminal ganglia (TGs) of mice, and are retained in latently infected ganglia. Here we demonstrate that CD8(+) T cells that are present in the TGs at the time of excision can maintain HSV-1 in a latent state in sensory neurons in ex vivo TG cultures. Latently infected neurons expressed viral genome and some expressed HSV-1 immediate early and early proteins, but did not produce HSV-1 late proteins or infectious virions. Addition of anti-CD8alpha monoclonal antibody 5 d after culture initiation induced HSV-1 reactivation, as demonstrated by production of viral late proteins and infectious virions. Thus, CD8(+) T cells can prevent HSV-1 reactivation without destroying the infected neurons. We propose that when the intrinsic capacity of neurons to inhibit HSV-1 reactivation from latency is compromised, production of HSV-1 immediate early and early proteins might activate CD8(+) T cells aborting virion production.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/growth & development , Neurons, Afferent/virology , Trigeminal Ganglion/virology , Animals , Cells, Cultured , Eye Infections, Viral/immunology , Female , Mice , Mice, Inbred BALB C , Trigeminal Ganglion/cytology , Virus Activation/immunology , Virus Latency/immunology
10.
Oncogene ; 38(24): 4886, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31068665

ABSTRACT

The final sentence of the Acknowledgements should be as follows: This work was supported by grants from Instituto de Salud Carlos III (BA15/00092), Spanish Ministry of Economy and Competitiveness/EU-ERDF (SAF2016-80626-R, SAF2013-49149-R, BFU2014-51672-REDC), Fundación CajaCanarias (AP2015/008) to RF, and the Australian National Health and Medical Research (NHMRC program grant to SRL and KKK (APP1017028).

11.
Sci Rep ; 9(1): 18771, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31801957

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Cancer Lett ; 439: 14-23, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30240587

ABSTRACT

RNA interference (RNAi) therapy is an emerging class of biopharmaceutical that has immense potential in cancer medicine. RNAi medicines are based on synthetic oligonucleotides that can suppress a target protein in tumour cells with high specificity. This review explores the attractive prospect of using RNAi as a radiosensitiser by targeting the DNA damage response. There are a multitude of molecular targets involved in the detection and repair of DNA damage that are suitable for this purpose. Recent developments in delivery technologies such nanoparticle carriers and conjugation strategies have allowed RNAi therapeutics to enter clinical trials in the treatment of cancer. With further progress, RNAi targeting of the DNA damage response may hold great promise in guiding radiation oncology into the era of precision medicine.


Subject(s)
DNA Damage , Neoplasms/radiotherapy , RNA Interference , Radiation Tolerance/genetics , Drug Delivery Systems/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Nanoparticles , Neoplasms/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
13.
Sci Rep ; 8(1): 73, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311580

ABSTRACT

Complex regulatory networks control epithelial-to-mesenchymal transition (EMT) but the underlying epigenetic control is poorly understood. Lysine-specific demethylase 1 (LSD1) is a key histone demethylase that alters the epigenetic landscape. Here we explored the role of LSD1 in global epigenetic regulation of EMT, cancer stem cells (CSCs), the tumour microenvironment, and therapeutic resistance in breast cancer. LSD1 induced pan-genomic gene expression in networks implicated in EMT and selectively elicits gene expression programs in CSCs whilst repressing non-CSC programs. LSD1 phosphorylation at serine-111 (LSD1-s111p) by chromatin anchored protein kinase C-theta (PKC-θ), is critical for its demethylase and EMT promoting activity and LSD1-s111p is enriched in chemoresistant cells in vivo. LSD1 couples to PKC-θ on the mesenchymal gene epigenetic template promotes LSD1-mediated gene induction. In vivo, chemotherapy reduced tumour volume, and when combined with an LSD1 inhibitor, abrogated the mesenchymal signature and promoted an innate, M1 macrophage-like tumouricidal immune response. Circulating tumour cells (CTCs) from metastatic breast cancer (MBC) patients were enriched with LSD1 and pharmacological blockade of LSD1 suppressed the mesenchymal and stem-like signature in these patient-derived CTCs. Overall, LSD1 inhibition may serve as a promising epigenetic adjuvant therapy to subvert its pleiotropic roles in breast cancer progression and treatment resistance.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Histone Demethylases/genetics , Transcriptional Activation , Tumor Microenvironment/genetics , Biomarkers , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Female , Gene Regulatory Networks , Histone Demethylases/metabolism , Histones/metabolism , Humans , Neoplastic Stem Cells/metabolism , Phenotype , Protein Transport , Signal Transduction
14.
Mol Cell Biol ; 21(15): 5214-22, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11438675

ABSTRACT

The checkpoint kinase Chk2 has a key role in delaying cell cycle progression in response to DNA damage. Upon activation by low-dose ionizing radiation (IR), which occurs in an ataxia telangiectasia mutated (ATM)-dependent manner, Chk2 can phosphorylate the mitosis-inducing phosphatase Cdc25C on an inhibitory site, blocking entry into mitosis, and p53 on a regulatory site, causing G(1) arrest. Here we show that the ATM-dependent activation of Chk2 by gamma- radiation requires Nbs1, the gene product involved in the Nijmegen breakage syndrome (NBS), a disorder that shares with AT a variety of phenotypic defects including chromosome fragility, radiosensitivity, and radioresistant DNA synthesis. Thus, whereas in normal cells Chk2 undergoes a time-dependent increased phosphorylation and induction of catalytic activity against Cdc25C, in NBS cells null for Nbs1 protein, Chk2 phosphorylation and activation are both defective. Importantly, these defects in NBS cells can be complemented by reintroduction of wild-type Nbs1, but neither by a carboxy-terminal deletion mutant of Nbs1 at amino acid 590, unable to form a complex with and to transport Mre11 and Rad50 in the nucleus, nor by an Nbs1 mutated at Ser343 (S343A), the ATM phosphorylation site. Chk2 nuclear expression is unaffected in NBS cells, hence excluding a mislocalization as the cause of failed Chk2 activation in Nbs1-null cells. Interestingly, the impaired Chk2 function in NBS cells correlates with the inability, unlike normal cells, to stop entry into mitosis immediately after irradiation, a checkpoint abnormality that can be corrected by introduction of the wild-type but not the S343A mutant form of Nbs1. Altogether, these findings underscore the crucial role of a functional Nbs1 complex in Chk2 activation and suggest that checkpoint defects in NBS cells may result from the inability to activate Chk2.


Subject(s)
DNA Damage , Enzyme Activation , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Checkpoint Kinase 2 , Enzyme Activation/radiation effects , Fibroblasts/metabolism , Gamma Rays , Gene Deletion , Humans , Immunoblotting , Microscopy, Fluorescence , Mitosis , Mutation , Phosphorylation , Phosphotransferases/metabolism , Precipitin Tests , Radiation, Ionizing , Time Factors , Transfection , cdc25 Phosphatases/metabolism
15.
Mol Biol Cell ; 12(5): 1199-213, 2001 May.
Article in English | MEDLINE | ID: mdl-11359916

ABSTRACT

Exposure to DNA-damaging agents triggers signal transduction pathways that are thought to play a role in maintenance of genomic stability. A key protein in the cellular processes of nucleotide excision repair, DNA recombination, and DNA double-strand break repair is the single-stranded DNA binding protein, RPA. We showed previously that the p34 subunit of RPA becomes hyperphosphorylated as a delayed response (4-8 h) to UV radiation (10-30 J/m(2)). Here we show that UV-induced RPA-p34 hyperphosphorylation depends on expression of ATM, the product of the gene mutated in the human genetic disorder ataxia telangiectasia (A-T). UV-induced RPA-p34 hyperphosphorylation was not observed in A-T cells, but this response was restored by ATM expression. Furthermore, purified ATM kinase phosphorylates the p34 subunit of RPA complex in vitro at many of the same sites that are phosphorylated in vivo after UV radiation. Induction of this DNA damage response was also dependent on DNA replication; inhibition of DNA replication by aphidicolin prevented induction of RPA-p34 hyperphosphorylation by UV radiation. We postulate that this pathway is triggered by the accumulation of aberrant DNA replication intermediates, resulting from DNA replication fork blockage by UV photoproducts. Further, we suggest that RPA-p34 is hyperphosphorylated as a participant in the recombinational postreplication repair of these replication products. Successful resolution of these replication intermediates reduces the accumulation of chromosomal aberrations that would otherwise occur as a consequence of UV radiation.


Subject(s)
DNA Repair/genetics , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ultraviolet Rays , Amino Acid Sequence , Aphidicolin/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Fractionation , Cell Line , Culture Media, Serum-Free , DNA Damage , DNA Replication/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/radiation effects , Enzyme Inhibitors/pharmacology , Humans , Immunoblotting , Molecular Sequence Data , Peptide Mapping , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Protein A , Tumor Suppressor Proteins , Xeroderma Pigmentosum/genetics
16.
Oncogene ; 36(7): 1012-1022, 2017 02 16.
Article in English | MEDLINE | ID: mdl-27568981

ABSTRACT

F-box proteins in conjunction with Skp1, Cul1 and Rbx1 generate SCF complexes that are responsible for the ubiquitination of proteins, leading to their activation or degradation. Here we show that the F-box protein FBXO31 is required for normal mitotic progression and genome stability due to its role in regulating FOXM1 levels during the G2/M transition. FBXO31-depleted cells undergo a transient delay in mitosis due to an activated spindle checkpoint concomitant with an increase in lagging chromosomes and anaphase bridges. FBXO31 regulates mitosis in part by controlling the levels of FOXM1, a transcription factor and master regulator of mitosis. FBXO31 specifically interacts with FOXM1 during the G2/M transition, resulting in FOXM1 ubiquitination and degradation. FBXO31 depletion results in increased expression of FOXM1 transcriptional targets and mimics the FOXM1 overexpression. In contrast, co-depletion of FBXO31 and FOXM1 restores the genomic instability phenotype but not the delay in mitosis, indicating that FBXO31 probably has additional mitotic substrates. Thus, FBXO31 is the first described negative regulator of FOXM1 during the G2/M transition.


Subject(s)
Cell Division/genetics , F-Box Proteins/metabolism , Forkhead Box Protein M1/metabolism , G2 Phase/genetics , Genomic Instability , Mitosis/genetics , Tumor Suppressor Proteins/metabolism , F-Box Proteins/genetics , Forkhead Box Protein M1/genetics , HEK293 Cells , HeLa Cells , Humans , Tumor Suppressor Proteins/genetics
17.
Oncogene ; 36(33): 4802-4809, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28288134

ABSTRACT

Correct control of DNA replication is crucial to maintain genomic stability in dividing cells. Inappropriate re-licensing of replicated origins is associated with chromosomal instability (CIN), a hallmark of cancer progression that at the same time provides potential opportunities for therapeutic intervention. Geminin is a critical inhibitor of the DNA replication licensing factor Cdt1. To properly achieve its functions, Geminin levels are tightly regulated through the cell cycle by ubiquitin-dependent proteasomal degradation, but the de-ubiquitinating enzymes (DUBs) involved had not been identified. Here we report that DUB3 and USP7 control human Geminin. Overexpression of either DUB3 or USP7 increases Geminin levels through reduced ubiquitination. Conversely, depletion of DUB3 or USP7 reduces Geminin levels, and DUB3 knockdown increases re-replication events, analogous to the effect of Geminin depletion. In exploring potential clinical implications, we found that USP7 and Geminin are strongly correlated in a cohort of invasive breast cancers (P<1.01E-08). As expected, Geminin expression is highly prognostic. Interestingly, we found a non-monotonic relationship between USP7 and breast cancer-specific survival, with both very low or high levels of USP7 associated with poor outcome, independent of estrogen receptor status. Altogether, our data identify DUB3 and USP7 as factors that regulate DNA replication by controlling Geminin protein stability, and suggest that USP7 may be involved in Geminin dysregulation during breast cancer progression.


Subject(s)
Breast Neoplasms/enzymology , Cell Cycle Proteins/antagonists & inhibitors , Endopeptidases/metabolism , Geminin/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Chromosomal Instability , DNA Replication/physiology , Disease Progression , Endopeptidases/genetics , HEK293 Cells , Humans , Kaplan-Meier Estimate , Neoplasm Invasiveness , Prognosis , Protein Stability , RNA, Small Interfering/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination
18.
Mucosal Immunol ; 10(2): 520-530, 2017 03.
Article in English | MEDLINE | ID: mdl-27461178

ABSTRACT

Mucosal antigen-specific CD4 T-cell responses to intestinal pathogens remain incompletely understood. Here we examined the CD4 T-cell response after oral infection with an internalin A 'murinized' Listeria monocytogenes (Lm). Oral Lm infection induced a robust endogenous listeriolysin O (LLO)-specific CD4 T-cell response with distinct phenotypic and functional characteristics in the intestine. Circulating LLO-specific CD4 T cells transiently expressed the 'gut-homing' integrin α4ß7 and accumulated in the intestinal lamina propria and epithelium where they were maintained independent of interleukin (IL)-15. The majority of intestinal LLO-specific CD4 T cells were CD27- Ly6C- and CD69+ CD103- while the lymphoid LLO-specific CD4 T cells were heterogeneous based on CD27 and Ly6C expression and predominately CD69-. LLO-specific effector CD4 T cells transitioned into a long-lived memory population that phenotypically resembled their parent effectors and displayed hallmarks of residency. In addition, intestinal effector and memory CD4 T cells showed a predominant polyfunctional Th1 profile producing IFNγ, TNFα, and IL-2 at high levels with minimal but detectable levels of IL-17A. Depletion of CD4 T cells in immunized mice led to elevated bacterial burden after challenge infection highlighting a critical role for memory CD4 T cells in controlling intestinal intracellular pathogens.


Subject(s)
Immunologic Memory , Intestinal Mucosa/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Th1 Cells/immunology , Administration, Oral , Animals , Antigens, CD/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Toxins/immunology , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Heat-Shock Proteins/immunology , Hemolysin Proteins/immunology , Integrin alpha4/metabolism , Integrin beta Chains/metabolism , Intestinal Mucosa/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Lymphocyte Homing/metabolism
20.
Oncogene ; 36(46): 6490-6500, 2017 11 16.
Article in English | MEDLINE | ID: mdl-28759041

ABSTRACT

The critical role of calcium signalling in processes related to cancer cell proliferation and invasion has seen a focus on pharmacological inhibition of overexpressed ion channels in specific cancer subtypes as a potential therapeutic approach. However, despite the critical role of calcium in cell death pathways, pharmacological activation of overexpressed ion channels has not been extensively evaluated in breast cancer. Here we define the overexpression of transient receptor potential vanilloid 4 (TRPV4) in a subgroup of breast cancers of the basal molecular subtype. We also report that pharmacological activation of TRPV4 with GSK1016790A reduced viability of two basal breast cancer cell lines with pronounced endogenous overexpression of TRPV4, MDA-MB-468 and HCC1569. Pharmacological activation of TRPV4 produced pronounced cell death through two mechanisms: apoptosis and oncosis in MDA-MB-468 cells. Apoptosis was associated with PARP-1 cleavage and oncosis was associated with a rapid decline in intracellular ATP levels, which was a consequence of, rather than the cause of, the intracellular ion increase. TRPV4 activation also resulted in reduced tumour growth in vivo. These studies define a novel therapeutic strategy for breast cancers that overexpress specific calcium permeable plasmalemmal ion channels with available selective pharmacological activators.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , TRPV Cation Channels/genetics , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Immunoblotting , Leucine/analogs & derivatives , Leucine/pharmacology , Mice, Inbred BALB C , Mice, Nude , Necrosis/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL