scholarly journals Genomic instability is an early event driving chromatin reorganization and escape from oncogene-induced senescence

2020 ◽  
Author(s):  
C Zampetidis ◽  
P Galanos ◽  
A Angelopoulou ◽  
Y Zhu ◽  
T Karamitros ◽  
...  

SUMMARYOncogene-induced senescence (OIS) is an inherent and important tumor suppressor mechanism. However, if not timely removed via immune surveillance, senescent cells will also present a detrimental side. Although this has mostly been attributed to the senescence-associated-secretory-phenotype (SASP) of these cells, we recently proposed that “escape” from the senescent state represents another unfavorable outcome. Here, we exploit genomic and functional data from a prototypical human epithelial cell model carrying an inducible CDC6 oncogene to identify an early-acquired recurrent chromosomal inversion, which harbors a locus encoding the circadian transcription factor BHLHE40. This inversion alone suffices for BHLHE40 activation upon CDC6 induction and for driving cell cycle re-entry and malignant transformation. In summary, we now provide strong evidence in support of genomic instability underlying “escape” from oncogene-induced senescence.HIGHLIGHTSOncogene driven error-prone repair produces early genetic lesions allowing escape from senescenceCells escaping oncogene-induced senescence display mutational signatures observed in cancer patientsA single recurrent inversion harboring a circadian TF gene suffices for bypassing oncogene-induced senescenceChromatin loop and compartment remodeling support the “escape” transcriptional program

1995 ◽  
Vol 15 (8) ◽  
pp. 4249-4259 ◽  
Author(s):  
A M Yahanda ◽  
J M Bruner ◽  
L A Donehower ◽  
R S Morrison

Loss or mutation of p53 is thought to be an early event in the malignant transformation of many human astrocytic tumors. To better understand the role of p53 in their growth and transformation, we developed a model employing cultured neonatal astrocytes derived from mice deficient in one (p53 +/-) or both (p53 -/-) p53 alleles, comparing them with wild-type (p53 +/+) cells. Studies of in vitro and in vivo growth and transformation were performed, and flow cytometry and karyotyping were used to correlate changes in growth with genomic instability. Early-passage (EP) p53 -/- astrocytes achieved higher saturation densities and had more rapid growth than EP p53 +/- and +/+ cells. The EP p53 -/- cells were not transformed, as they were unable to grow in serum-free medium or in nude mice. With continued passaging, p53 -/- cells exhibited a multistep progression to a transformed phenotype. Late-passage p53 -/- cells achieved saturation densities 50 times higher than those of p53 +/+ cells and formed large, well-vascularized tumors in nude mice. p53 +/- astrocytes exhibited early loss of the remaining wild-type p53 allele and then evolved in a manner phenotypically similar to p53 -/- astrocytes. In marked contrast, astrocytes retaining both wild-type p53 alleles never exhibited a transformed phenotype and usually senesced after 7 to 10 passages. Dramatic alterations in ploidy and karyotype occurred and were restricted to cells deficient in wild-type p53 following repeated passaging. The results of these studies suggest that loss of wild-type p53 function promotes genomic instability, accelerated growth, and malignant transformation in astrocytes.


Cells ◽  
2019 ◽  
Vol 8 (5) ◽  
pp. 414 ◽  
Author(s):  
Nazmul Huda ◽  
Yan Xu ◽  
Alison M. Bates ◽  
Deborah A. Rankin ◽  
Nagarajan Kannan ◽  
...  

Telomere dysfunction has been strongly implicated in the initiation of genomic instability and is suspected to be an early event in the carcinogenesis of human solid tumors. Recent findings have established the presence of telomere fusions in human breast and prostate malignancies; however, the onset of this genomic instability mechanism during progression of other solid cancers is not well understood. Herein, we explored telomere dynamics in patient-derived epithelial ovarian cancers (OC), a malignancy characterized by multiple distinct subtypes, extensive molecular heterogeneity, and widespread genomic instability. We discovered a high frequency of telomere fusions in ovarian tumor tissues; however, limited telomere fusions were detected in normal adjacent tissues or benign ovarian samples. In addition, we found relatively high levels of both telomerase activity and hTERT expression, along with anaphase bridges in tumor tissues, which were notably absent in adjacent normal ovarian tissues and benign lesions. These results suggest that telomere dysfunction may occur early in ovarian carcinogenesis and, importantly, that it may play a critical role in the initiation and progression of the disease. Recognizing telomere dysfunction as a pervasive feature of this heterogeneous malignancy may facilitate the future development of novel diagnostic tools and improved methods of disease monitoring and treatment.


2020 ◽  
Vol 10 ◽  
Author(s):  
Peng Liu ◽  
Qinghe Tang ◽  
Miaomiao Chen ◽  
Wenjian Chen ◽  
Yanli Lu ◽  
...  

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. The lack of effective targeted drugs has become a challenge on treating HCC patients. Cellular senescence is closely linked to the occurrence, development, and therapy of tumor. Induction of cellular senescence and further activation of immune surveillance provides a new strategy to develop HCC targeted drugs, that is, senescence-induced therapy for HCC. Precancerous hepatocytes or HCC cells can be induced into senescent cells, subsequently producing senescence-associated secretory phenotype (SASP) factors. SASP factors recruit and activate various types of immune cells, including T cells, NK cells, macrophages, and their subtypes, which carry out the role of immune surveillance and elimination of senescent cells, ultimately preventing the occurrence of HCC or inhibiting the progression of HCC. Specific interventions in several checkpoints of senescence-mediated therapy will make positive contributions to suppress tumorigenesis and progression of HCC, for instance, by applying small molecular compounds to induce cellular senescence or selecting cytokines/chemokines to activate immunosurveillance, supplementing adoptive immunocytes to remove senescent cells, and screening chemical drugs to induce apoptosis of senescent cells or accelerate clearance of senescent cells. These interventional checkpoints become potential chemotherapeutic targets in senescence-induced therapy for HCC. In this review, we focus on the frontiers of senescence-induced therapy and discuss senescent characteristics of hepatocytes during hepatocarcinogenesis as well as the roles and mechanisms of senescent cell induction and clearance, and cellular senescence-related immunosurveillance during the formation and progression of HCC.


2020 ◽  
Vol 21 (15) ◽  
pp. 5193
Author(s):  
Elizabeth Terry-Kantor ◽  
Arati Tripathi ◽  
Thibaut Imberdis ◽  
Zachary M. LaVoie ◽  
Gary P. H. Ho ◽  
...  

Genetic and biochemical evidence attributes neuronal loss in Parkinson’s disease (PD) and related brain diseases to dyshomeostasis of the 14 kDa protein α-synuclein (αS). There is no consensus on how αS exerts toxicity. Explanations range from disturbed vesicle biology to proteotoxicity caused by fibrillar aggregates. To probe these mechanisms further, robust cellular toxicity models are needed, but their availability is limited. We previously reported that a shift from dynamic multimers to monomers is an early event in αS dyshomeostasis, as caused by familial PD (fPD)-linked mutants such as E46K. Excess monomers accumulate in round, lipid-rich inclusions. Engineered αS ‘3K’ (E35K+E46K+E61K) amplifies E46K, causing a PD-like, L-DOPA-responsive motor phenotype in transgenic mice. Here, we present a cellular model of αS neurotoxicity after transducing human neuroblastoma cells to express yellow fluorescent protein (YFP)-tagged αS 3K in a doxycycline-dependent manner. αS-3K::YFP induction causes pronounced growth defects that accord with cell death. We tested candidate compounds for their ability to restore growth, and stearoyl-CoA desaturase (SCD) inhibitors emerged as a molecule class with growth-restoring capacity, but the therapeutic window varied among compounds. The SCD inhibitor MF-438 fully restored growth while exerting no apparent cytotoxicity. Our αS bioassay will be useful for elucidating compound mechanisms, for pharmacokinetic studies, and for compound/genetic screens.


Endocrinology ◽  
2014 ◽  
Vol 155 (9) ◽  
pp. 3315-3328 ◽  
Author(s):  
Kandice L. Tessneer ◽  
Robert M. Jackson ◽  
Beth A. Griesel ◽  
Ann Louise Olson

Abstract Glucose transporter isoform 4 (GLUT4) is the insulin-responsive glucose transporter mediating glucose uptake in adipose and skeletal muscle. Reduced GLUT4 translocation from intracellular storage compartments to the plasma membrane is a cause of peripheral insulin resistance. Using a chronic hyperinsulinemia (CHI)-induced cell model of insulin resistance and Rab5 mutant overexpression, we determined these manipulations altered endosomal sorting of GLUT4, thus contributing to the development of insulin resistance. We found that CHI induced insulin resistance in 3T3-L1 adipocytes by retaining GLUT4 in a Rab5-activity-dependent compartment that is unable to equilibrate with the cell surface in response to insulin. Furthermore, CHI-mediated retention of GLUT4 in this non-insulin-responsive compartment impaired filling of the transferrin receptor (TfR)-positive and TfR-negative insulin-responsive storage compartments. Our data suggest that hyperinsulinemia may inhibit GLUT4 by chronically maintaining GLUT4 in the Rab5 activity-dependent endosomal pathway and impairing formation of the TfR-negative and TfR-positive insulin-responsive GLUT4 pools. This model suggests that an early event in the development of insulin-resistant glucose transport in adipose tissue is to alter the intracellular localization of GLUT4 to a compartment that does not efficiently equilibrate with the cell surface when insulin levels are elevated for prolonged periods of time.


Author(s):  
Maria Ryaboshapkina ◽  
Kevin Saitoski ◽  
Ghaith M. Hamza ◽  
Andrew F. Jarnuczak ◽  
Claire Berthault ◽  
...  

ABSTRACTEarly diabetes research is hampered by limited availability, variable quality and instability of human pancreatic islets in culture. Little is known about the human β cell secretome, and recent studies question translatability of rodent β cell secretory profiles. Here, we verify representativeness of EndoC-βH1, one of the most widely used human β cell lines, as a translational human β cell model based on omics and characterize the EndoC-βH1 secretome. We profiled EndoC-βH1 cells using RNA-seq, Data Independent Acquisition (DIA) and Tandem Mass Tag proteomics of cell lysate. Omics profiles of EndoC-βH1 cells were compared to human β cells and insulinomas. Secretome composition was assessed by DIA proteomics. Agreement between EndoC-βH1 cells and primary adult human β cells was ~90% for global omics profiles as well as for β cell markers, transcription factors and enzymes. Discrepancies in expression were due to elevated proliferation rate of EndoC-βH1 cells compared to adult β cells. Consistently, similarity was slightly higher with benign non-metastatic insulinomas. EndoC-βH1 secreted 671 proteins in untreated baseline state and 3,278 proteins when stressed with non-targeting control siRNA, including known β cell hormones INS, IAPP, and IGF2. Further, EndoC-βH1 secreted proteins known to generate bioactive peptides such as granins and enzymes required for production of bioactive peptides. Unexpectedly, exosomes appeared to be a major mode of secretion in EndoC-βH1 cells. We believe that secretion of exosomes and bioactive peptides warrant further investigation with specialized proteomics workflows in future studies.Graphical abstractHighlightsWe validate EndoC-βH1 as a translational human β cell model using omics.We present the first unbiased proteomics composition of human β cell line secretome.The secretome of human β cells is more extensive than previously thought.Untreated cells secreted 671 proteins and stressed cells secreted 3,278 proteins.Secretion of exosomes and bioactive peptides constitute directions of future research.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 200-200
Author(s):  
Feyruz V. Rassool ◽  
Terry J. Gaymes ◽  
Nicola Brady ◽  
Nicholas Lea ◽  
Marika Pla ◽  
...  

Abstract Genomic instability is the driving force of disease progression to frank leukemia. Evidence suggests that aberrant repair of double strand breaks (DSB) by non homologous end-joining (NHEJ), a major repair pathway in mammalian cells, can lead to chromosomal instability and cancer. We previously reported significantly increased error-prone NHEJ in preleukemic syndromes, and a variety of myeloid malignancies, and demonstrated that these cells harbor constitutive DNA damage. We postulated that increased NHEJ misrepair may be a response to the increased DNA damage. Here, we have studied a mouse model for myeloproliferative/myelodysplastic syndrome (MPD/MDS) to determine whether the frequency of DNA damage and aberrant NHEJ repair may be an indicator for genomic instability as the disease progresses. Transgenic mice bearing mutant NRAS and BCL-2 driven by the MRP8 promoter, which directs expression of the transgene to committed myeloid progenitors and neutrophils, have a relatively mild phenotype with an increase of immature neutrophils. The BCL2 mice have an increase in marrow blasts, but have normal blood counts. Transgenic mice harboring both mutant NRAS and BCL2 genes results in a disease phenotype morphologically resembling human late MDS (FAB subtypes refractory anemia with excess blasts (RAEB), RAEB in transformation (RAEBt) or chronic myelomonocytic leukaemia (CMML)) with increased marrow blasts. We show that the bone marrow and spleen from the NRAS and BCL2 mice demonstrate an increase in the frequency of NHEJ misrepair activity, compared with normal (FVBN) mice (NRAS: 7.6% vs 3.7%, BCL2: 6.5% vs 3.7%, n=3). Strikingly, the NRAS +BCL2 double transgenic mice show a large and significant increase in NHEJ misrepair activity (19.02%, n=3, p<0.001), compared with controls and single transgenics. Using an immunofluorescence-based assay for DNA damage, dependent on BrdU incorporation, we find that the magnitude of DNA damage mirrors NHEJ activity. Chromatin fibers from both NRAS and BCL2 mice demonstrate an increase in the frequency of DNA damage, compared to normal mice (NRAS: 35% vs 8%, mean [n=3]), (BCL2 22% vs 8%, [n=3]). However, this damage increases even further in RAS +BCL2 mice (62% vs NRAS/BCL2 28%, [FVBN] 8%, n=3, p<0.001). This DNA damage co-localizes with the variant histone γH2AX, a key protein in the repair of DSB. DNA damage and γH2AX also co-localize with the NHEJ protein Ku86 emphasizing that DNA damage is linked to repair by NHEJ in situ. Given that activated RAS produces increased reactive oxygen species (ROS), an established source for DSB, we considered whether ROS accounted for some of this DNA damage. We find that cells from transgenic mice show an increase (up to 2-fold) in ROS, compared with controls. The same is true for FDCP1 murine cells transduced with NRAS and BCL2, and treatment with the antioxidant, N-acetyl cysteine results in an up to 50% decrease in ROS, DNA damage and concomitant NHEJ misrepair activity. Our data suggest that increased DNA damage and error-prone repair may be a platform for the creation of increased genomic instability with disease progression in MPD/MDS in mice. Decreased DNA damage and error-prone repair with antioxidant treatment suggests a mechanism for the amelioration of the activities that drive disease progression.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1806-1806
Author(s):  
Sabina Sevcikova ◽  
Fedor Kryukov ◽  
Elena Dementyeva ◽  
Kubiczkova Lenka ◽  
Pavel Nemec ◽  
...  

Abstract Abstract 1806 Background: Multiple myeloma (MM) is a malignancy of differentiated B-lymphocytes characterized by accumulation of clonal plasma cells (PCs) in the bone marrow. Genome of malignant PCs is extremely unstable and characterized by complex combination of structural and numerical abnormalities. The basis of genomic instability underlying MM is still unclear. Centrosome amplification (CA) is present in about a third of MM cases and may represent a mechanism leading to genomic instability in myeloma. We have previously shown that CA is present in pre-plasma cells stages of B-cell development. Aims: The objective of our study was to evaluate changes in expression of chosen key genes involved in centrosome duplication process in both B and PCs, including their possible changes during MM progression. Materials and Methods: In total, 56 patients were evaluated for this study. Patients' characteristics were as follows: males/females 38/37, median age 69 years (range 43–83 years). Newly diagnosed (27/56) and relapsed (29/56) patients were included in this study; most of them had advanced stage of MM (DS III n=48; ISS II/III n=45). qRT-PCR (51 PCs and 32 B cell samples) was performed on a chosen set of mitotic genes, according to their role in normal centrosome duplication process. Results: Expression of genes involving in centrosome duplication process was studied in B cells of MM patients and healthy donors. Group of MM patients showed significant increase in relative quantification coefficient R in the following genes: AURKA, AURKB, CCNB1, CCNB2, HMMR, PLK4, TACC3 and TUBG1. It is notable that TUBG1 was approximately 330-times overexpressed in MM B cells vs healthy donors. Expression of studied genes was significantly different in B and PCs populations of MM patients: AURKB and TACC3 were upregulated in B cells while CENT2 and TUBG1 were upregulated in PCs. Analyses of gene expression in newly diagnosed and relapsed patients showed significant changes of the following genes: AURKA and TACC3 were 2-times upregulated in PCs of relapsed patients; no differences were detected in population of B cells. Conclusion: Considering revealed changes in B cells of myeloma patients, we suspect early damage in cell-cycle regulating mechanism. This hypothesis correlates with our previous findings of CA in MM B cells. We anticipate that dynamic accumulation of showed changes plays a role in final transformation of malignant PCs. We assume that centrosomal disruption likely contributes to accumulation of genomic abnormalities in tumor cells during MM progression which can be proven in future long-term study. Acknowledgment: This study was supported by grants NT11154, NS10207, MSM0021622434 Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document