scholarly journals A role for keratin 17 during DNA damage response and tumor initiation

2021 ◽  
Vol 118 (13) ◽  
pp. e2020150118
Author(s):  
Raji R. Nair ◽  
Joshua Hsu ◽  
Justin T. Jacob ◽  
Christopher M. Pineda ◽  
Ryan P. Hobbs ◽  
...  

High levels of the intermediate filament protein keratin 17 (K17) are associated with poor prognoses for several human carcinomas. Studies in mouse models have shown that K17 expression is positively associated with growth, survival, and inflammation in skin and that lack of K17 delays onset of tumorigenesis. K17 occurs in the nucleus of human and mouse tumor keratinocytes where it impacts chromatin architecture, gene expression, and cell proliferation. We report here that K17 is induced following DNA damage and promotes keratinocyte survival. The presence of nuclear K17 is required at an early stage of the double-stranded break (DSB) arm of the DNA damage and repair (DDR) cascade, consistent with its ability to associate with key DDR effectors, including γ-H2A.X, 53BP1, and DNA-PKcs. Mice lacking K17 or with attenuated K17 nuclear import showed curtailed initiation in a two-step skin carcinogenesis paradigm. The impact of nuclear-localized K17 on DDR and cell survival provides a basis for the link between K17 induction and poor clinical outcomes for several human carcinomas.

Cancers ◽  
2020 ◽  
Vol 12 (7) ◽  
pp. 1717 ◽  
Author(s):  
Sara Sofia Deville ◽  
Anne Vehlow ◽  
Sarah Förster ◽  
Ellen Dickreuter ◽  
Kerstin Borgmann ◽  
...  

The treatment resistance of cancer cells is a multifaceted process in which DNA repair emerged as a potential therapeutic target. DNA repair is predominantly conducted by nuclear events; yet, how extra-nuclear cues impact the DNA damage response is largely unknown. Here, using a high-throughput RNAi-based screen in three-dimensionally-grown cell cultures of head and neck squamous cell carcinoma (HNSCC), we identified novel focal adhesion proteins controlling DNA repair, including the intermediate filament protein, synemin. We demonstrate that synemin critically regulates the DNA damage response by non-homologous end joining repair. Mechanistically, synemin forms a protein complex with DNA-PKcs through its C-terminal tail domain for determining DNA repair processes upstream of this enzyme in an ATM-dependent manner. Our study discovers a critical function of the intermediate filament protein, synemin in the DNA damage response, fundamentally supporting the concept of cytoarchitectural elements as co-regulators of nuclear events.


Metabolites ◽  
2021 ◽  
Vol 11 (8) ◽  
pp. 513
Author(s):  
Grace H. Yang ◽  
Danielle A. Fontaine ◽  
Sukanya Lodh ◽  
Joseph T. Blumer ◽  
Avtar Roopra ◽  
...  

Transcription factor 19 (TCF19) is a gene associated with type 1 diabetes (T1DM) and type 2 diabetes (T2DM) in genome-wide association studies. Prior studies have demonstrated that Tcf19 knockdown impairs β-cell proliferation and increases apoptosis. However, little is known about its role in diabetes pathogenesis or the effects of TCF19 gain-of-function. The aim of this study was to examine the impact of TCF19 overexpression in INS-1 β-cells and human islets on proliferation and gene expression. With TCF19 overexpression, there was an increase in nucleotide incorporation without any change in cell cycle gene expression, alluding to an alternate process of nucleotide incorporation. Analysis of RNA-seq of TCF19 overexpressing cells revealed increased expression of several DNA damage response (DDR) genes, as well as a tightly linked set of genes involved in viral responses, immune system processes, and inflammation. This connectivity between DNA damage and inflammatory gene expression has not been well studied in the β-cell and suggests a novel role for TCF19 in regulating these pathways. Future studies determining how TCF19 may modulate these pathways can provide potential targets for improving β-cell survival.


2019 ◽  
Vol 48 (1) ◽  
pp. 68
Author(s):  
Michael E. Devitt ◽  
Robert Dreicer

<p>The aim of this article is to review the current role of genomic testing in the risk, prognosis, and treatment of genitourinary malignancies. The authors selected guidelines, publications, and abstracts relevant to the current and emerging role of genomics in genitourinary cancers. The risk of developing genitourinary cancer can be stratified based on genomic data. Prostate cancer has the strongest degree of heritability, with <em>BRCA1/2 </em>and <em>HOXB13 </em>mutations playing a role in familial disease. Genomic data is on the verge of informing treatment decisions across genitourinary cancers. mCRPC has diverse genomic alterations that represent potential therapeutic targets, including alterations in the AR pathway, DNA damage and repair pathways, cell cycle pathways, PI3K pathway, and Wnt signaling. Genomic alterations in clear cell renal cell carcinoma can inform prognosis and mutations in mTOR pathways predict response to mTOR inhibitors. Urothelial carcinoma can be classified into different subtypes based on gene expression profiling, which provides prognostic information and predicts response to chemotherapy and immunotherapy. Specific mutations have been identified that predict response to therapy including <em>ERCC2 </em>mutations and cisplatin, DNA damage and repair mutations and checkpoint inhibitors, and <em>FGFR3 </em>mutations and FGFR tyrosine kinase inhibitors such as erdafitinib.</p><p><strong>Conclusion. </strong>Genitourinary malignancies have not felt the impact of genomic data as greatly as other cancer types. The majority of benefit lies in identifying patients at high risk of genitourinary cancer. Fortunately, breakthroughs are on the horizon that will result in a greater incorporation of genomic information into treatment decisions for patients with genitourinary cancer.</p>


2018 ◽  
Vol 5 (1) ◽  
pp. 141-164 ◽  
Author(s):  
Matthew D. Weitzman ◽  
Amélie Fradet-Turcotte

Viral DNA genomes have limited coding capacity and therefore harness cellular factors to facilitate replication of their genomes and generate progeny virions. Studies of viruses and how they interact with cellular processes have historically provided seminal insights into basic biology and disease mechanisms. The replicative life cycles of many DNA viruses have been shown to engage components of the host DNA damage and repair machinery. Viruses have evolved numerous strategies to navigate the cellular DNA damage response. By hijacking and manipulating cellular replication and repair processes, DNA viruses can selectively harness or abrogate distinct components of the cellular machinery to complete their life cycles. Here, we highlight consequences for viral replication and host genome integrity during the dynamic interactions between virus and host.


2014 ◽  
Vol 32 (15_suppl) ◽  
pp. 4015-4015 ◽  
Author(s):  
Richard C. Turkington ◽  
Laura A. Hill ◽  
Damian McManus ◽  
Stephen McQuaid ◽  
Ken Arthur ◽  
...  

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1570-1570
Author(s):  
Roisin M McAvera ◽  
Jonathan J Morgan ◽  
Ken I Mills ◽  
Lisa J Crawford

Abstract Introduction Chromosomal instability is a hallmark of Multiple Myeloma (MM), with most patients displaying cytogenetic abnormalities which can arise due to DNA damage response (DDR) defects. TRIM33 is an E3 ligase and transcriptional co-repressor located on chromosome 1p13.2, a region frequently deleted in MM. Previous studies have shown that TRIM33 plays a role in the DDR and can regulate chromosomal stability, but its precise function remains unknown. In this study we investigated the impact of TRIM33 loss in MM on genomic stability and DDR pathways and whether this could be exploited therapeutically. Methods The CoMMpass dataset (IA15 release) was screened to identify patients with copy number (CN) loss of TRIM33 and this was correlated with overall survival (OS) and structural variants. TRIM33 shRNA knockdown models were established in JJN3 and U266 cells. The effect on DDR signalling was determined by western blotting and immunofluorescence. The Selleckchem DNA Damage/Repair Compound Library was screened on the JJN3 model in a high-throughput manner using the CellTox™ Green cytotoxicity assay. Validation of selected compounds was performed using CellTiter® Glo viability assay or clonogenic assays. Combination indices (CI) were calculated using CompuSyn software. Results Data on CN, OS and structural variants were available for 730 newly diagnosed MM patients and of these, 69 (9.5%) were identified to have a CN loss of TRIM33. These patients have poorer OS compared to those without TRIM33 loss (52.3 months vs 72.6 months; p&lt;0.0001). Moreover, they exhibit a significantly higher median number of structural variants (deletions, duplications, inversions, and translocations; 38 vs 26; p&lt;0.0001), indicative of increased chromosomal instability. Our data in MM cell lines has shown that TRIM33 is rapidly recruited to chromatin within 5 minutes of induced DNA damage. TRIM33 knockdown led to an increase in 53BP1 foci formation and endogenous γH2AX (P&lt;0.001) indicating unrepaired DNA double-strand breaks (DSBs) typical of a DDR defect. In response to these DSBs both ATM and ATR kinases were activated as demonstrated by increased pKAP1 Ser824 and pCHK1 Ser345 respectively (p&lt;0.001). Additionally, we observed a reduction in RAD51 (p&lt;0.05) indicative of a potential defect in the DSB repair pathway homologous recombination (HR). To identify therapeutic vulnerabilities relating to TRIM33 loss, we performed a high-throughput screen to assess sensitivity to 160 unique DNA damaging compounds. TRIM33 knockdown cells exhibited increased sensitivity to 27 compounds across a range of drug classes. Additional studies confirmed that compared to control cells, TRIM33 knockdown sensitized cells to the PARP inhibitor Olaparib and ATR inhibitors BAY-1895344 and VE-821. Further investigation with VE-821 demonstrated that whilst treatment induced PARP cleavage and DSBs in both control and knockdown cells within 48 hours, knockdown cells exhibited significantly more pCHK1 Ser345 inhibition (p&lt;0.01). Furthermore, combining VE-821 with bortezomib yielded synergistic effects in TRIM33 knockdown cells across a range of doses (CI range 0.57-0.9) while no synergy was observed in control cells (CI&gt;1 for all combinations). Conclusion We have identified a subset of MM patients with TRIM33 loss who display high-risk disease characterized by chromosomal abnormalities and defective DDR. Alongside this we have identified PARP and ATR inhibitors as therapeutic vulnerabilities in cell line models of TRIM33 loss. Moreover, we demonstrate that ATR inhibition increases the efficacy of bortezomib in TRIM33 knockdown cells. Further investigation into these compounds could lead to novel therapies for patients with TRIM33 loss. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 394 (10) ◽  
pp. 1325-1331 ◽  
Author(s):  
Maike Sauer ◽  
Katrin S. Reiners ◽  
Hinrich P. Hansen ◽  
Andreas Engert ◽  
Stephan Gasser ◽  
...  

Abstract Evasion of apoptosis is a hallmark of cancer cells. Inhibitor of apoptosis proteins (IAPs) act as endogenous inhibitors of programmed cell death and are overexpressed in several tumors including Hodgkin lymphoma (HL). Preclinical studies indicate antitumor activity of IAP antagonists and clinical studies in hematological malignancies are underway. Here, we investigate the impact of the small molecule IAP antagonist LCL161 on HL cell lines. Although the antagonist caused rapid degradation of cIAP1 leading to TNFα secretion, LCL161 did not promote apoptosis significantly. However, LCL161 induced expression of MICA and MICB, ligands for the activating immune receptor NKG2D, and enhanced the susceptibility of HL cells to NKG2D-dependent lysis by NK cells. MICA/B upregulation was dependent on activation of the DNA damage response upon LCL161 treatment. Taken together, we demonstrate a novel link between IAP inhibition, DNA damage and immune recognition.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e20037-e20037
Author(s):  
Shaopeng Zheng ◽  
Jin Xia ◽  
Yunfang Yu ◽  
Fanjun Zeng ◽  
Luyu Huang ◽  
...  

e20037 Background: In recent years, there has been a better understanding of ways to use DNA damage and repair (DDR) mechanisms to improve overall sensitivity and/or overcome resistance to traditional DNA damage treatments. The DDR network is quite complex and highly dynamic with as many as 450 proteins integral to the DNA repair. The current study is to identify the key dysregulated genes and its related pathways especially DDR pathways in early progression of lung adenocarcinoma. Methods: TCGA dataset of lung adenocarcinoma (LUAD) including 59 healthy lung tissues and 517 tumor tissues was utilized to detect the differentially expressed mRNAs and lncRNAs. Gene ontology (GO) analysis was conducted with DAVID, while Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of differentially expressed genes was performed using gene set enrichment analysis (GSEA) methods. Results: 41 lncRNAs and 2,047 mRNAs were screened out to be differentially expressed in LUAD. Besides, 38 lncRNAs and 1,801 mRNAs were found to be differentially expressed in T1 stage LUAD. The homologous recombination repair (HRR) pathway was found to be significantly up-regulated in LUAD, with four genes in this pathway up-regulated. In these genes of HRR pathway, PPP4R4 and RAD54L were recognized to be significantly differential expressed in T1 stage, compared with T2 stage and T3 stage, which were putative biomarkers of early stage LUAD. The survival analysis revealed that the expression of RAD54L was significantly related to the survival rate of patients with tumor of T1 stage. Conclusions: HRR pathway was up-regulated in lung adenocarcinoma, in which the expression of PPP4R4 and RAD54L were found to be tumor stage specific and RAD54L was related with survival rate of T1 stage patients. This study provided a further insight into the mechanism of the progression in early stage lung adenocarcinoma.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 437-437
Author(s):  
William G. Wierda ◽  
Kumudha Balakrishnan ◽  
Alessandra Ferrajoli ◽  
Susan O'Brien ◽  
Jan A. Burger ◽  
...  

Abstract Abstract 437 Chemoimmunotherapy (such as fludarabine, cyclophosphamide, and rituximab) is highly effective CLL therapy; adding novel agents or replacing standard with more effective agents could improve outcomes. Bendamustine (B) is a potent alkylating agent that induces DNA damage and repair response. Marked DNA damage response (H2AX phosphorylation) was seen with activation of p53 protein and PUMA, and cell death when fludarabine was combined with bendamustine in vitro. Base on this, we are conducting a phase I/II trial of escalating doses of bendamustine at 20 (n=6), 30 (n=17), 40 (n=6), or 50 (n=6) mg/m2 on D1, 2, 3 with fludarabine 20 mg/m2 administered prior to bendamustine on D2 & 3. Rituximab 375–500 mg/m2 was given D3. Courses were repeated each 28 days for 6 planned courses to assess the safety and tolerability, clinical efficacy, and pharmacodynamics (PD) in previously treated CLL. Response assessment (IWCLL 2008 criteria) was after 3 courses and end of treatment; bone marrow residual disease was assessed by 4-color flow. We previously reported (ASH 2011) that no MTD was identified in phase I and identified bendamustine 30 mg/m2 as safe for phase II expansion. We now report on efficacy of this FBR regimen in 35 pts treated in phase I & II who have response data available. The median age was 62 yrs; number of prior treatments was 3 (1-6); and number of FBR courses was 3 (1-6). Dose-reduction after course 1 occurred in 10/35 pts. Responses are shown (Table) and were seen across all dose levels. Time-to-event endpoints will be presented. Pts had high-risk features: median b-2 microglobulin was 4.1 (1.8–10.4); 18/35 were Rai stage III-IV; 23/29 were ZAP70+; 23/29 had unmutated IGHV; and FISH identified 3 pts with del17p and 13 with del11q. Myelosuppression was the most common treatment-related toxicity considering all courses given (n=106). Grade(G) 3&G4 neutropenia occurred in 26&29% of courses, respectively; thrombocytopenia G3&G4 occurred in 14&9% of courses, respectively; and anemia G3&G4 occurred in 15&2% of courses, respectively. There were no treatment-related deaths. To test fludarabine triphosphate-mediated mitigation of DNA repair response induced by bendamustine, on D1 bendamustine was infused alone and on D2, fludarabine was administered 2 hours prior to second bendamustine infusion; circulating CLL cells from 11 pts at different bendamustine doses (3 at 20, 3 at 30, 3 at 40, and 2 at 50 mg/m2) were evaluated. Phosphorylation of histone 2A variant × (H2AX) was used as damage response marker. There was heterogeneity in extent of DNA damage response elicited after first bendamustine infusion. Considering basal phosphorylation level in the pretreatment sample on D1 as 1.0, the H2AX phosphorylation at D1-6hr (bendamustine alone) ranged between 0.2–8 (n=11). Median intracellular fludarabine triphosphate level at the start of bendamustine infusion was 12 μM (range 5–21 μM). This was sufficient to increase H2AX phosphorylation in all 11 pts tested. At the end of D2-4hr (bendamustine combined with fludarabine) the range was between 0.9–22 and remained the same on D2-6hr. In 1 pt sample, the phosphorylation persisted at 26 until D3-pretreatement, while in others it ranged between 3–12. Consistent with H2AX data, molecular markers of DNA damage response showed activation of ATM measured in 5 pt samples as ser1981 phosphorylation and phosphorylation of p53 at ser15. In parallel, there was a decrease in anti-apoptotic proteins Mcl-1 and Bcl-2 at the end of D2-6hr; however protein levels were retained on D3. In conclusion, this FBR regimen was tolerated up to the highest evaluated bendamustine dose; efficacy was demonstrated in previously treated pts with CLL. DNA damage and repair response biomarkers validated the hypothesis that fludarabine triphosphate inhibited bendamustine-induced DNA repair resulting in increased or sustained DNA damage. We continue to extend the clinical and PD investigations in phase II. Table Characteristic n % CR/CRi % OR %MRD Neg All Pts 35 26 71 11 Rai Stage III-IV 18 17 67 11 0-II 17 35 76 12 No. Prior Rx >2 19 16 58 5 1-2 16 38 88 19 B2M 34 mg/l 19 11 53 11 <4 mg/l 16 44 94 13 FISH 17p del 3 33 100 0 11q del 13 23 69 0 +12 7 57 86 43 None 3 0 33 33 13q del 3 0 100 0 IGHV Unmutated 23 26 70 9 Mutated 6 33 100 33 ZAP70 Positive 23 22 70 13 Negative 6 17 83 17 CD38 (>7%) Positive 26 23 73 8 Negative 8 38 75 25 CR, complete remission; CRi, CR with incomplete recovery of cytopenias; OR, overall response; MRD, minimal residual disease. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document