Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Sci Rep ; 14(1): 7270, 2024 03 27.
Article in English | MEDLINE | ID: mdl-38538606

ABSTRACT

Cancer risk is associated with the widely debated measure body mass index (BMI). Fat mass and fat-free mass measurements from bioelectrical impedance may further clarify this association. The UK Biobank is a rare resource in which bioelectrical impedance and BMI data was collected on ~ 500,000 individuals. Using this dataset, a comprehensive analysis using regression, principal component and genome-wide genetic association, provided multiple levels of evidence that increasing whole body fat (WBFM) and fat-free mass (WBFFM) are both associated with increased post-menopausal breast cancer risk, and colorectal cancer risk in men. WBFM was inversely associated with prostate cancer. We also identified rs615029[T] and rs1485995[G] as associated in independent analyses with both PMBC (p = 1.56E-17 and 1.78E-11) and WBFFM (p = 2.88E-08 and 8.24E-12), highlighting splice variants of the intriguing long non-coding RNA CUPID1 (LINC01488) as a potential link between PMBC risk and fat-free mass.


Subject(s)
Body Composition , Neoplasms , Male , Humans , Body Composition/genetics , Body Mass Index , Genetic Predisposition to Disease , Neoplasms/etiology , Neoplasms/genetics , Electric Impedance
2.
Genome Biol ; 24(1): 128, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37221612

ABSTRACT

BACKGROUND: Therapy resistance in cancer is often driven by a subpopulation of cells that are temporarily arrested in a non-proliferative G0 state, which is difficult to capture and whose mutational drivers remain largely unknown. RESULTS: We develop methodology to robustly identify this state from transcriptomic signals and characterise its prevalence and genomic constraints in solid primary tumours. We show that G0 arrest preferentially emerges in the context of more stable, less mutated genomes which maintain TP53 integrity and lack the hallmarks of DNA damage repair deficiency, while presenting increased APOBEC mutagenesis. We employ machine learning to uncover novel genomic dependencies of this process and validate the role of the centrosomal gene CEP89 as a modulator of proliferation and G0 arrest capacity. Lastly, we demonstrate that G0 arrest underlies unfavourable responses to various therapies exploiting cell cycle, kinase signalling and epigenetic mechanisms in single-cell data. CONCLUSIONS: We propose a G0 arrest transcriptional signature that is linked with therapeutic resistance and can be used to further study and clinically track this state.


Subject(s)
Genomics , Neoplasms , Humans , Cell Cycle Checkpoints , Cell Cycle , Mutagenesis
4.
Front Genome Ed ; 4: 932434, 2022.
Article in English | MEDLINE | ID: mdl-35865001

ABSTRACT

CRISPR/Cas9, base editors and prime editors comprise the contemporary genome editing toolbox. Many studies have optimized the use of CRISPR/Cas9, as the original CRISPR genome editing system, in substituting single nucleotides by homology directed repair (HDR), although this remains challenging. Studies describing modifications that improve editing efficiency fall short of isolating clonal cell lines or have not been validated for challenging loci or cell models. We present data from 95 transfections using a colony forming and an immortalized cell line comparing the effect on editing efficiency of donor template modifications, concentration of components, HDR enhancing agents and cold shock. We found that in silico predictions of guide RNA efficiency correlated poorly withactivity in cells. Using NGS and ddPCR we detected editing efficiencies of 5-12% in the transfected populations which fell to 1% on clonal cell line isolation. Our data demonstrate the variability of CRISPR efficiency by cell model, target locus and other factors. Successful genome editing requires a comparison of systems and modifications to develop the optimal protocol for the cell model and locus. We describe the steps in this process in a flowchart for those embarking on genome editing using any system and incorporate validated HDR-boosting modifications for those using CRISPR/Cas9.

5.
Cancer Epidemiol Biomarkers Prev ; 31(7): 1450-1459, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35477182

ABSTRACT

BACKGROUND: The genetic factors that modulate risk for developing lung cancer have not been fully defined. Here, we sought to determine the prevalence and clinical significance of germline pathogenic/likely pathogenic variants (PV) in patients with advanced lung cancer. METHODS: We studied clinical and tumor characteristics of germline PV in 5,118 patients who underwent prospective genomic profiling using paired tumor-normal tissue samples in 468 cancer genes. RESULTS: Germline PV in high/moderate-penetrance genes were observed in 222 (4.3%) patients; of these, 193 patients had PV in DNA damage repair (DDR) pathway genes including BRCA2 (n = 54), CHEK2 (n = 30), and ATM (n = 26) that showed high rate of biallelic inactivation in tumors. BRCA2 heterozygotes with lung adenocarcinoma were more likely to be never smokers and had improved survival compared with noncarriers. Fourteen patients with germline PV in lung cancer predisposing genes (TP53, EGFR, BAP1, and MEN1) were diagnosed at younger age compared with noncarriers, and of tumor suppressors, 75% demonstrated biallelic inactivation in tumors. A significantly higher proportion of germline PV in high/moderate-penetrance genes were detected in high-risk patients who had either a family history of any cancer, multiple primary tumors, or early age at diagnosis compared with unselected patients (10.5% vs. 4.1%; P = 1.7e-04). CONCLUSIONS: These data underscore the biological and clinical importance of germline mutations in highly penetrant DDR genes as a risk factor for lung cancer. IMPACT: The family members of lung cancer patients harboring PV in cancer predisposing genes should be referred for genetic counseling and may benefit from proactive surveillance.


Subject(s)
Genetic Predisposition to Disease , Lung Neoplasms , Germ Cells , Germ-Line Mutation , Humans , Lung Neoplasms/genetics , Prospective Studies
6.
Cancer Res ; 81(7): 1667-1680, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33558336

ABSTRACT

Insights into oncogenesis derived from cancer susceptibility loci (SNP) hold the potential to facilitate better cancer management and treatment through precision oncology. However, therapeutic insights have thus far been limited by our current lack of understanding regarding both interactions of these loci with somatic cancer driver mutations and their influence on tumorigenesis. For example, although both germline and somatic genetic variation to the p53 tumor suppressor pathway are known to promote tumorigenesis, little is known about the extent to which such variants cooperate to alter pathway activity. Here we hypothesize that cancer risk-associated germline variants interact with somatic TP53 mutational status to modify cancer risk, progression, and response to therapy. Focusing on a cancer risk SNP (rs78378222) with a well-documented ability to directly influence p53 activity as well as integration of germline datasets relating to cancer susceptibility with tumor data capturing somatically-acquired genetic variation provided supportive evidence for this hypothesis. Integration of germline and somatic genetic data enabled identification of a novel entry point for therapeutic manipulation of p53 activities. A cluster of cancer risk SNPs resulted in increased expression of prosurvival p53 target gene KITLG and attenuation of p53-mediated responses to genotoxic therapies, which were reversed by pharmacologic inhibition of the prosurvival c-KIT signal. Together, our results offer evidence of how cancer susceptibility SNPs can interact with cancer driver genes to affect cancer progression and identify novel combinatorial therapies. SIGNIFICANCE: These results offer evidence of how cancer susceptibility SNPs can interact with cancer driver genes to affect cancer progression and present novel therapeutic targets.


Subject(s)
Drug Resistance, Neoplasm/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Pharmacological/metabolism , Carcinogenesis/genetics , Case-Control Studies , Cell Line, Tumor , Disease Progression , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Germ-Line Mutation/physiology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation, Missense , Neoplasms/diagnosis , Neoplasms/drug therapy , Polymorphism, Single Nucleotide/physiology , Prognosis , Risk Factors , Signal Transduction/genetics , Treatment Outcome
7.
J Med Genet ; 58(6): 392-399, 2021 06.
Article in English | MEDLINE | ID: mdl-32591342

ABSTRACT

BACKGROUND: Height and other anthropometric measures are consistently found to associate with differential cancer risk. However, both genetic and mechanistic insights into these epidemiological associations are notably lacking. Conversely, inherited genetic variants in tumour suppressors and oncogenes increase cancer risk, but little is known about their influence on anthropometric traits. METHODS: By integrating inherited and somatic cancer genetic data from the Genome-Wide Association Study Catalog, expression Quantitative Trait Loci databases and the Cancer Gene Census, we identify SNPs that associate with different cancer types and differential gene expression in at least one tissue type, and explore the potential pleiotropic associations of these SNPs with anthropometric traits through SNP-wise association in a cohort of 500,000 individuals. RESULTS: We identify three regulatory SNPs for three important cancer genes, FANCA, MAP3K1 and TP53 that associate with both anthropometric traits and cancer risk. Of particular interest, we identify a previously unrecognised strong association between the rs78378222[C] SNP in the 3' untranslated region (3'-UTR) of TP53 and both increased risk for developing non-melanomatous skin cancer (OR=1.36 (95% 1.31 to 1.41), adjusted p=7.62E-63), brain malignancy (OR=3.12 (2.22 to 4.37), adjusted p=1.43E-12) and increased standing height (adjusted p=2.18E-24, beta=0.073±0.007), lean body mass (adjusted p=8.34E-37, beta=0.073±0.005) and basal metabolic rate (adjusted p=1.13E-31, beta=0.076±0.006), thus offering a novel genetic link between these anthropometric traits and cancer risk. CONCLUSION: Our results clearly demonstrate that heritable variants in key cancer genes can associate with both differential cancer risk and anthropometric traits in the general population, thereby lending support for a genetic basis for linking these human phenotypes.


Subject(s)
Body Weights and Measures , Neoplasms/genetics , Oncogenes , Polymorphism, Single Nucleotide , Adult , Aged , Anthropometry , Cohort Studies , Female , Genetic Linkage , Genetic Pleiotropy , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Middle Aged , Quantitative Trait Loci , Quantitative Trait, Heritable , Risk Assessment
8.
Br J Cancer ; 122(8): 1231-1241, 2020 04.
Article in English | MEDLINE | ID: mdl-32147670

ABSTRACT

BACKGROUND: Genome-wide association studies (GWASs) have enriched the fields of genomics and drug development. Adrenocortical carcinoma (ACC) is a rare cancer with a bimodal age distribution and inadequate treatment options. Paediatric ACC is frequently associated with TP53 mutations, with particularly high incidence in Southern Brazil due to the TP53 p.R337H (R337H) germline mutation. The heterogeneous risk among carriers suggests other genetic modifiers could exist. METHODS: We analysed clinical, genotype and gene expression data derived from paediatric ACC, R337H carriers, and adult ACC patients. We restricted our analyses to single nucleotide polymorphisms (SNPs) previously identified in GWASs to associate with disease or human traits. RESULTS: A SNP, rs971074, in the alcohol dehydrogenase 7 gene significantly and reproducibly associated with allelic differences in ACC age-of-onset in both cohorts. Patients homozygous for the minor allele were diagnosed up to 16 years earlier. This SNP resides in a gene involved in the retinoic acid (RA) pathway and patients with differing levels of RA pathway gene expression in their tumours associate with differential ACC progression. CONCLUSIONS: These results identify a novel genetic component to ACC development that resides in the retinoic acid pathway, thereby informing strategies to develop management, preventive and therapeutic treatments for ACC.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , Genes, p53 , Polymorphism, Single Nucleotide , Tretinoin/physiology , Adolescent , Adrenal Cortex Neoplasms/epidemiology , Adrenocortical Carcinoma/epidemiology , Age Factors , Age of Onset , Alcohol Dehydrogenase/genetics , Child , Child, Preschool , Female , Genome-Wide Association Study , Humans , Incidence , Infant , Male
9.
Proc Natl Acad Sci U S A ; 116(35): 17470-17479, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31395738

ABSTRACT

The most frequently mutated protein in human cancer is p53, a transcription factor (TF) that regulates myriad genes instrumental in diverse cellular outcomes including growth arrest and cell death. Cell context-dependent p53 modulation is critical for this life-or-death balance, yet remains incompletely understood. Here we identify sequence signatures enriched in genomic p53-binding sites modulated by the transcription cofactor iASPP. Moreover, our p53-iASPP crystal structure reveals that iASPP displaces the p53 L1 loop-which mediates sequence-specific interactions with the signature-corresponding base-without perturbing other DNA-recognizing modules of the p53 DNA-binding domain. A TF commonly uses multiple structural modules to recognize its cognate DNA, and thus this mechanism of a cofactor fine-tuning TF-DNA interactions through targeting a particular module is likely widespread. Previously, all tumor suppressors and oncoproteins that associate with the p53 DNA-binding domain-except the oncogenic E6 from human papillomaviruses (HPVs)-structurally cluster at the DNA-binding site of p53, complicating drug design. By contrast, iASPP inhibits p53 through a distinct surface overlapping the E6 footprint, opening prospects for p53-targeting precision medicine to improve cancer therapy.


Subject(s)
DNA/genetics , DNA/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Repressor Proteins/metabolism , Response Elements , Tumor Suppressor Protein p53/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , DNA/chemistry , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Nucleotide Motifs , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/metabolism , Protein Binding , Protein Conformation , Repressor Proteins/chemistry , Structure-Activity Relationship , Tumor Suppressor Protein p53/chemistry
11.
Curr Opin Genet Dev ; 48: 112-120, 2018 02.
Article in English | MEDLINE | ID: mdl-29216518

ABSTRACT

Cancer development involves the stepwise accumulation of genetic lesions that overcome the normal regulatory pathways that prevent unconstrained cell division and tissue growth. Identification of the genetic changes that cause cancer has long been the subject of intensive study, leading to the identification of several RNA-binding proteins (RBPs) linked to cancer. Cross-reference of the complement of RBPs recently identified by RNA interactome capture with cancer-associated genes and biological processes led to the identification of a set of 411 proteins with potential implications in cancer biology. These involve a broad spectrum of cellular processes including response to stress, metabolism and cell adhesion. Future studies should aim to understand these proteins and their connection to cancer from an RNA-centred perspective, holding the promise of new mechanistic understanding of cancer formation and novel approaches to diagnosis and treatment.


Subject(s)
Neoplasms/genetics , RNA-Binding Proteins/metabolism , Animals , Carcinogenesis , Humans , Neoplasms/metabolism , RNA-Binding Proteins/genetics
12.
Cancer Res ; 77(6): 1250-1260, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28254861

ABSTRACT

Accurate assessment of TP53 gene status in sporadic tumors and in the germline of individuals at high risk of cancer due to Li-Fraumeni Syndrome (LFS) has important clinical implications for diagnosis, surveillance, and therapy. Genomic data from more than 20,000 cancer genomes provide a wealth of information on cancer gene alterations and have confirmed TP53 as the most commonly mutated gene in human cancer. Analysis of a database of 70,000 TP53 variants reveals that the two newly discovered exons of the gene, exons 9ß and 9γ, generated by alternative splicing, are the targets of inactivating mutation events in breast, liver, and head and neck tumors. Furthermore, germline rearrange-ments in intron 1 of TP53 are associated with LFS and are frequently observed in sporadic osteosarcoma. In this context of constantly growing genomic data, we discuss how screening strategies must be improved when assessing TP53 status in clinical samples. Finally, we discuss how TP53 alterations should be described by using accurate nomenclature to avoid confusion in scientific and clinical reports. Cancer Res; 77(6); 1250-60. ©2017 AACR.


Subject(s)
Genetic Variation/genetics , Neoplasms/genetics , Practice Guidelines as Topic/standards , Quality Control , Tumor Suppressor Protein p53/genetics , Humans , Neoplasms/diagnosis , Neoplasms/therapy , Validation Studies as Topic
13.
Genes Dev ; 30(20): 2297-2309, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27898394

ABSTRACT

Angiogenesis, the fundamental process by which new blood vessels form from existing ones, depends on precise spatial and temporal gene expression within specific compartments of the endothelium. However, the molecular links between proangiogenic signals and downstream gene expression remain unclear. During sprouting angiogenesis, the specification of endothelial cells into the tip cells that lead new blood vessel sprouts is coordinated by vascular endothelial growth factor A (VEGFA) and Delta-like ligand 4 (Dll4)/Notch signaling and requires high levels of Notch ligand DLL4. Here, we identify MEF2 transcription factors as crucial regulators of sprouting angiogenesis directly downstream from VEGFA. Through the characterization of a Dll4 enhancer directing expression to endothelial cells at the angiogenic front, we found that MEF2 factors directly transcriptionally activate the expression of Dll4 and many other key genes up-regulated during sprouting angiogenesis in both physiological and tumor vascularization. Unlike ETS-mediated regulation, MEF2-binding motifs are not ubiquitous to all endothelial gene enhancers and promoters but are instead overrepresented around genes associated with sprouting angiogenesis. MEF2 target gene activation is directly linked to VEGFA-induced release of repressive histone deacetylases and concurrent recruitment of the histone acetyltransferase EP300 to MEF2 target gene regulatory elements, thus establishing MEF2 factors as the transcriptional effectors of VEGFA signaling during angiogenesis.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/physiology , Gene Expression Regulation, Developmental , MEF2 Transcription Factors/metabolism , Neovascularization, Physiologic/genetics , Animals , Cells, Cultured , Embryo, Nonmammalian , Endothelial Cells/enzymology , Enhancer Elements, Genetic/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MEF2 Transcription Factors/chemistry , MEF2 Transcription Factors/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neovascularization, Pathologic/genetics , Protein Interaction Domains and Motifs , Retina/embryology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
14.
Clin Cancer Res ; 22(24): 6069-6077, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27283965

ABSTRACT

PURPOSE: Although pancreatic ductal adenocarcinoma (PDAC) is an aggressive tumor, like other common cancers, it displays a wide range of biology. However, at present, there are no reliable tests to predict patients' cancer-specific outcomes and guide personalized treatment decisions. In this study, we aim to identify such biomarkers in resectable PDAC by studying SNPs in the CD44 gene, which drives the progression of pancreatic cancer. EXPERIMENTAL DESIGN: A total of 348 PDAC patients from three independent cohorts [Switzerland, Germany, The Cancer Genome Atlas (TCGA)] who underwent pancreatic resection are included in the study. Information on the haplotype structure of the CD44 gene is obtained using 1000 Genomes Project data, and the genotypes of the respective tagging SNPs are determined. Cox proportional hazards models are utilized to analyze the impact of SNP genotype on patients' survival. RESULTS: We identify an SNP in the CD44 gene (SNPrs187115) that independently associates with allelic differences in prognosis in all study cohorts. Specifically, in 121 Swiss patients, we observe an up to 2.38-fold (P = 0.020) difference in tumor-related death between the genotypes of SNPrs187115 We validate those results in both the German (HR = 2.32, P = 0.044, 101 patients) and the TCGA cohort (HR = 2.36, P = 0.044, 126 patients). CONCLUSIONS: CD44 SNPrs187115 can serve as a novel biomarker readily available at the time of PDAC diagnosis that identifies patients at risk for faster tumor progression and guide personalized treatment decisions. It has the potential to significantly expand the pool of patients that would benefit from tumor resection. Clin Cancer Res; 22(24); 6069-77. ©2016 AACR.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Hyaluronan Receptors/genetics , Pancreatic Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/pathology , Cohort Studies , Disease Progression , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Prognosis , Proportional Hazards Models , Young Adult , Pancreatic Neoplasms
15.
Cell Rep ; 15(4): 830-842, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27149848

ABSTRACT

The NRF2/sMAF protein complex regulates the oxidative stress response by occupying cis-acting enhancers containing an antioxidant response element (ARE). Integrating genome-wide maps of NRF2/sMAF occupancy with disease-susceptibility loci, we discovered eight polymorphic AREs linked to 14 highly ranked disease-risk SNPs in individuals of European ancestry. Among these SNPs was rs242561, located within a regulatory region of the MAPT gene (encoding microtubule-associated protein Tau). It was consistently occupied by NRF2/sMAF in multiple experiments and its strong-binding allele associated with higher mRNA levels in cell lines and human brain tissue. Induction of MAPT transcription by NRF2 was confirmed using a human neuroblastoma cell line and a Nrf2-deficient mouse model. Most importantly, rs242561 displayed complete linkage disequilibrium with a highly protective allele identified in multiple GWASs of progressive supranuclear palsy, Parkinson's disease, and corticobasal degeneration. These observations suggest a potential role for NRF2/sMAF in tauopathies and a possible role for NRF2 pathway activators in disease prevention.

16.
Nat Rev Cancer ; 16(4): 251-65, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27009395

ABSTRACT

Decades of research have shown that mutations in the p53 stress response pathway affect the incidence of diverse cancers more than mutations in other pathways. However, most evidence is limited to somatic mutations and rare inherited mutations. Using newly abundant genomic data, we demonstrate that commonly inherited genetic variants in the p53 pathway also affect the incidence of a broad range of cancers more than variants in other pathways. The cancer-associated single nucleotide polymorphisms (SNPs) of the p53 pathway have strikingly similar genetic characteristics to well-studied p53 pathway cancer-causing somatic mutations. Our results enable insights into p53-mediated tumour suppression in humans and into p53 pathway-based cancer surveillance and treatment strategies.


Subject(s)
Genetic Predisposition to Disease/genetics , Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Protein p53/genetics , Genome, Human , Humans , Mutation
17.
Cancer Res ; 75(4): 698-708, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25649770

ABSTRACT

TP53 gene mutation is associated with poor prognosis in breast cancer, but additional biomarkers that can further refine the impact of the p53 pathway are needed to achieve clinical utility. In this study, we evaluated a role for the HDMX-S/FL ratio as one such biomarker, based on its association with other suppressor mutations that confer worse prognosis in sarcomas, another type of cancer that is surveilled by p53. We found that HDMX-S/FL ratio interacted with p53 mutational status to significantly improve prognostic capability in patients with breast cancer. This biomarker pair offered prognostic utility that was comparable with a microarray-based prognostic assay. Unexpectedly, the utility tracked independently of DNA-damaging treatments and instead with different tumor metastasis potential. Finally, we obtained evidence that this biomarker pair might identify patients who could benefit from anti-HDM2 strategies to impede metastatic progression. Taken together, our work offers a p53 pathway marker, which both refines our understanding of the impact of p53 activity on prognosis and harbors potential utility as a clinical tool.


Subject(s)
Breast Neoplasms/genetics , Lymphatic Metastasis/genetics , Nuclear Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle Proteins , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis/pathology , Mutation , Neoplasm Staging , Tumor Suppressor Protein p53/genetics
18.
Int J Radiat Biol ; 90(10): 903-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24882388

ABSTRACT

PURPOSE: Although there are considerable data on mechanisms of radiation-induced apoptosis in vitro and in animal models, little is known about functional variation in these pathways in humans. We sought to develop a tractable system to evaluate this. MATERIALS AND METHODS: Peripheral blood mononuclear cells were isolated from 90 healthy volunteers, divided into two aliquots, one irradiated with a 5 Gy dose and the other sham-treated (0 Gy), and assessed for damage-induced apoptosis after 24 hours. To investigate reproducibility, 10 individuals spanning the entire radiation-induced apoptotic range were tested three times each, with 3-6 months between replicates. RESULTS: We observed surprising heterogeneity in apoptosis among individuals, ranging from 21-62%. Biological replicates from a single individual, however, were completely concordant, suggesting the variability observed across individuals is not the result of stochastic or short-term effects. We found significantly higher radiation-induced apoptosis in males than in females (Mean: 41.0% vs. 30.7%; p < 3.5 × 10(-7)). Moreover, advancing age was associated with decreasing radiation-induced apoptosis in males (p = 0.01) but not females (p = 0.82). CONCLUSIONS: Our results provide evidence that the function of cellular pathways crucial for stress-induced apoptosis varies by sex and could decline with age in humans.


Subject(s)
Aging/physiology , Apoptosis/physiology , Apoptosis/radiation effects , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/radiation effects , Models, Biological , Adult , Cells, Cultured , Computer Simulation , Female , Humans , Leukocytes, Mononuclear/cytology , Male , Middle Aged , Radiation Dosage , Reference Values , Sex Factors , Young Adult
20.
Cell ; 155(2): 410-22, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120139

ABSTRACT

The ability of p53 to regulate transcription is crucial for tumor suppression and implies that inherited polymorphisms in functional p53-binding sites could influence cancer. Here, we identify a polymorphic p53 responsive element and demonstrate its influence on cancer risk using genome-wide data sets of cancer susceptibility loci, genetic variation, p53 occupancy, and p53-binding sites. We uncover a single-nucleotide polymorphism (SNP) in a functional p53-binding site and establish its influence on the ability of p53 to bind to and regulate transcription of the KITLG gene. The SNP resides in KITLG and associates with one of the largest risks identified among cancer genome-wide association studies. We establish that the SNP has undergone positive selection throughout evolution, signifying a selective benefit, but go on to show that similar SNPs are rare in the genome due to negative selection, indicating that polymorphisms in p53-binding sites are primarily detrimental to humans.


Subject(s)
Genome-Wide Association Study , Polymorphism, Single Nucleotide , Response Elements , Stem Cell Factor/genetics , Testicular Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Proliferation , Genetic Predisposition to Disease , Humans , Male , Mice , Selection, Genetic , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...