Kathleen Klotz-Noack’s research while affiliated with Charité Universitätsmedizin Berlin and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (9)


Figure 4. MAPK activity is linked to CRC cell differentiation states. A Gene expression of LGR5 and EPHB2, along activity gradients of LGR5-ISC or MAPK target gene signatures. B Cell state distribution of SCN-aberrant CRC cells along gradients of LGR5-ISC or MAPK transcriptional signatures, as in A. C Cell state distribution of SCN-aberrant CRC tumor cells along MAPK signature activity, as in B, per tumor. Correlation between cell state distributions and MAPK target gene was calculated using Pearson's r. For correlations and significances, see Table EV6. Color code as in Fig 4B. D UMAP representations of single-cell transcriptomes derived from P009T or P013T organoids, after MAPK blockade using MEK or combined MEK and EGFR inhibition. Color codes are treatment conditions or expression strength of signature, as indicated. Dashed line in P013T UMAP roughly separates control (DMSO) and MEK/EGFR inhibitor-treated cells.
Mitogen‐activated protein kinase activity drives cell trajectories in colorectal cancer
  • Article
  • Full-text available

August 2021

·

156 Reads

·

71 Citations

EMBO Molecular Medicine

·

·

·

[...]

·

In colorectal cancer, oncogenic mutations transform a hierarchically organized and homeostatic epithelium into invasive cancer tissue lacking visible organization. We sought to define transcriptional states of colorectal cancer cells and signals controlling their development by performing single-cell transcriptome analysis of tumors and matched non-cancerous tissues of twelve colorectal cancer patients. We defined patient-overarching colorectal cancer cell clusters characterized by differential activities of oncogenic signaling pathways such as mitogen-activated protein kinase and oncogenic traits such as replication stress. RNA metabolic labeling and assessment of RNA velocity in patient-derived organoids revealed developmental trajectories of colorectal cancer cells organized along a mitogen-activated protein kinase activity gradient. This was in contrast to normal colon organoid cells developing along graded Wnt activity. Experimental targeting of EGFR-BRAF-MEK in cancer organoids affected signaling and gene expression contingent on predictive KRAS/BRAF mutations and induced cell plasticity overriding default developmental trajectories. Our results highlight directional cancer cell development as a driver of non-genetic cancer cell heterogeneity and re-routing of trajectories as a response to targeted therapy.

Download

Mutation-specific effects of NRAS oncogenes in colorectal cancer cells

December 2020

·

71 Reads

·

10 Citations

Advances in Biological Regulation

In colorectal cancer (CRC), the prevalence of NRAS mutations (5–9%) is inferior to that of KRAS mutations (40–50%). NRAS mutations feature lately during tumour progression and drive resistance to anti-EGFR therapy in KRAS wild-type tumours. To elucidate specific functions of NRAS mutations in CRC, we expressed doxycycline-inducible G12D and Q61K mutations in the CRC cell line Caco-2. A focused phospho-proteome analysis based on the Bio-Plex platform, which interrogated the activity of MAPK, PI3K, mTOR, STAT, p38, JNK and ATF2, did not reveal significant differences between Caco-2 cells expressing NRASG12D, NRASQ61K and KRASG12V. However, phenotypic read-outs were different. The NRAS Q61K mutation promoted anchorage-independent proliferation and tumorigenicity, similar to features driven by canonical KRAS mutations. In contrast, expression of NRASG12D resulted in reduced proliferation and apoptosis. At the transcriptome level, we saw upregulation of cytokines and chemokines. IL1A, IL11, CXCL8 (IL-8) and CCL20 exhibited enhanced secretion into the culture medium. In addition, RNA sequencing results indicated activation of the IL1-, JAK/STAT-, NFκB- and TNFα signalling pathways. These results form the basis for an NRASG12D-driven inflammatory phenotype in CRC.


SFPQ Depletion Is Synthetically Lethal with BRAFV600E in Colorectal Cancer Cells

September 2020

·

79 Reads

·

27 Citations

Cell Reports

Oncoproteins such as the BRAFV600E kinase endow cancer cells with malignant properties, but they also create unique vulnerabilities. Targeting of BRAFV600E-driven cytoplasmic signaling networks has proved ineffective, as patients regularly relapse with reactivation of the targeted pathways. We identify the nuclear protein SFPQ to be synthetically lethal with BRAFV600E in a loss-of-function shRNA screen. SFPQ depletion decreases proliferation and specifically induces S-phase arrest and apoptosis in BRAFV600E-driven colorectal and melanoma cells. Mechanistically, SFPQ loss in BRAF-mutant cancer cells triggers the Chk1-dependent replication checkpoint, results in decreased numbers and reduced activities of replication factories, and increases collision between replication and transcription. We find that BRAFV600E-mutant cancer cells and organoids are sensitive to combinations of Chk1 inhibitors and chemically induced replication stress, pointing toward future therapeutic approaches exploiting nuclear vulnerabilities induced by BRAFV600E.


Fig. 1 (See legend on next page.)
Fig. 2 Consistent growth reduction of MCM7-suppressed colorectal cancer cell lines. a Clonogenic assay quantification of SW480 (KRAS G12V ), HCT-8 (KRAS G13A ), HT-29 (BRAF V600E ), WiDr (BRAF V600E ). All cell lines showed decreased growth after doxycycline induction of MCM7.sh3 compared to the control shRNA (scrbl). Mean percentage of the relative area covered by cells ± SEM is shown (n = 3 per group). Student's t-test (two-sided); *p < 0.05; ***p < 0.001. b Clonogenic assay quantification of DLD1 KRAS(wt/−) and its parental cell line DLD1 KRAS(wt/G13D) . Both cell lines are sensitive to MCM7 suppression, but DLD1 cells without mutated KRAS become more resistant to low levels of MCM7. Mean percentage of the relative area covered by cells ± SEM is shown (n = 3 per group). Student's t-test (two-sided); NS = not significant; *p < 0.05; ****p < 0.0001. c Quantification of soft agar assays confirmed decreased anchorage-independent colony formation in KRAS mutated DLD1 and SW480 cells. Mean fold change of colonies ± SEM is shown (n = 6 per group). Student's t-test (two-sided); **p < 0.01; ****p < 0.0001.
Fig. 3 (See legend on next page.)
Fig. 4 (See legend on next page.)
Reduced replication origin licensing selectively kills KRAS-mutant colorectal cancer cells via mitotic catastrophe

July 2020

·

83 Reads

·

6 Citations

Cell Death and Disease

To unravel vulnerabilities of KRAS-mutant CRC cells, a shRNA-based screen specifically inhibiting MAPK pathway components and targets was performed in CaCo2 cells harboring conditional oncogenic KRASG12V. The custom-designed shRNA library comprised 121 selected genes, which were previously identified to be strongly regulated in response to MEK inhibition. The screen showed that CaCo2 cells expressing KRASG12V were sensitive to the suppression of the DNA replication licensing factor minichromosome maintenance complex component 7 (MCM7), whereas KRASwt CaCo2 cells were largely resistant to MCM7 suppression. Similar results were obtained in an isogenic DLD-1 cell culture model. Knockdown of MCM7 in a KRAS-mutant background led to replication stress as indicated by increased nuclear RPA focalization. Further investigation showed a significant increase in mitotic cells after simultaneous MCM7 knockdown and KRASG12V expression. The increased percentage of mitotic cells coincided with strongly increased DNA damage in mitosis. Taken together, the accumulation of DNA damage in mitotic cells is due to replication stress that remained unresolved, which results in mitotic catastrophe and cell death. In summary, the data show a vulnerability of KRAS-mutant cells towards suppression of MCM7 and suggest that inhibiting DNA replication licensing might be a viable strategy to target KRAS-mutant cancers.


Figure 6. Concurrent Inhibition of Compensatory Signaling Enhances RAFi/ERKi Growth Inhibition (A) Pa02C and Pa14C cells were treated with RAFi (0.3 mM), ERKi (0.04 mM), FRAX597 (PAKi, 1 mM), or MK2206 (AKTi, 0.6 mM) alone or in combination (slash indicates concurrent inhibition; plus sign indicates sequential inhibition, inhibitor addition after 72 h). Remaining cells were stained with crystal violet after a total of 5 days. Data are the mean average of two independent experiments. Error bars are shown as ± SEM. (B) Pa02C and Pa14C cells were treated as in (A) for a total of 5 days. Cell lysates were immunoblotted to determine the levels of the indicated proteins. Data are representative of two independent experiments.
Low-Dose Vertical Inhibition of the RAF-MEK-ERK Cascade Causes Apoptotic Death of KRAS Mutant Cancers

June 2020

·

209 Reads

·

79 Citations

Cell Reports

We address whether combinations with a pan-RAF inhibitor (RAFi) would be effective in KRAS mutant pancreatic ductal adenocarcinoma (PDAC). Chemical library and CRISPR genetic screens identify combinations causing apoptotic anti-tumor activity. The most potent combination, concurrent inhibition of RAF (RAFi) and ERK (ERKi), is highly synergistic at low doses in cell line, organoid, and rat models of PDAC, whereas each inhibitor alone is only cytostatic. Comprehensive mechanistic signaling studies using reverse phase protein array (RPPA) pathway mapping and RNA sequencing (RNA-seq) show that RAFi/ERKi induced insensitivity to loss of negative feedback and system failures including loss of ERK signaling, FOSL1, and MYC; shutdown of the MYC transcriptome; and induction of mesenchymal-to-epithelial transition. We conclude that low-dose vertical inhibition of the RAF-MEK-ERK cascade is an effective therapeutic strategy for KRAS mutant PDAC.


Abstract A20: Depletion of replication factor MCM7 is synthetically lethal to oncogenic KRAS expression

October 2017

·

13 Reads

Molecular Cancer Therapeutics

Colorectal cancer (CRC) is a substantial cause of death in the western world. With KRAS being one of the most frequently altered oncogenes in CRC, it is an obvious target for cancer therapy. But despite enormous efforts over the past three decades to target mutated RAS, not a single drug has made it to the clinic. In order to find vulnerabilities of KRAS mutant cells, we performed a shRNA-based screen. We used CaCo2 cells (wild type for KRAS/BRAF/PIK3CA) with doxycycline (DOX) inducible expression of the oncogenic KRASG12V as a model system. The custom-designed shRNA library comprised 121 selected genes encoding signaling proteins and transcription factors, which we have previously identified to be strongly up- or downregulated after treatment with MEK inhibitors. We found that CaCo2 cells expressing KRASG12V are highly sensitive to the depletion of the DNA replication licensing factor Minichromosome Maintenance Complex Component 7 (MCM7), whereas wild-type CaCo2 cells are resistant to MCM7 depletion. The screening result was verified with 3 independent shRNAs under anchorage dependent and independent conditions. Similar results were obtained with NRASG12D and in DLD1 (KRASwt/KRASG13D) and DLD1 (KRASwt/-) cells. The proposed molecular mechanism is RAS-induced genotoxic stress that cannot be resolved after MCM7 reduction. MCM7-containing complexes prime DNA for replication by binding to replication origins. However, only a few of the licensed origins are later used for DNA replication, while most origins stay dormant. The dormant origins are essential for resolving stalled replication forks after replicative stress. Unresolved stalled forks lead to DNA damage, which drives the cell into apoptosis. Further experiments will show the extent of DNA damage upon RAS induction and knock down of MCM7. Since DNA replication licensing is only required in actively dividing cells and cells with physiological RAS signaling seem to be resistant to partial depletion of MCM7, it might be an ideal target for treating RAS mutated cancers. Citation Format: Bastian Gastl, Kathleen Klotz-Noack, Bertram Klinger, Johannes Zuber, Nils Blüthgen, Reinhold Schäfer, Christine Sers. Depletion of replication factor MCM7 is synthetically lethal to oncogenic KRAS expression [abstract]. In: Proceedings of the AACR Precision Medicine Series: Opportunities and Challenges of Exploiting Synthetic Lethality in Cancer; Jan 4-7, 2017; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2017;16(10 Suppl):Abstract nr A20.


Abstract 5170: HDAC inhibitors and the mechanism of resistance in colorectal cancer: RAS and MYC - the partners in crime

July 2017

·

34 Reads

Cancer Research

Although histone deacetylase inhibitors (HDACi) are considered a promising novel therapeutic approach in the light of their potent tumour-selective effects, the use of these inhibitors for treatment of colorectal cancer (CRC) have thus far demonstrated limited success as a monotherapy. What this eventually boils down to is our incomplete understanding of the molecular mechanisms, the impact of oncogenes, and thus the key pathways through which HDACi affect tumour cell growth. To shed further light on this, the involvement of oncogenic RAS - a key driver of CRC, in determining the responsiveness to HDACi has been explored. By using cell line model systems harbouring conditional oncogenic NRAS, KRAS and HRAS, we uncovered an oncogenic RAS-dependent “safeguard” mechanism imposed in order to evade the cytotoxic effect of HDACi and therefore apoptosis. Characteristically, cells harbouring oncogenic RAS were observed to undergo a reversible senescence-like growth arrest in G2, allowing for re-entry into cell cycle following the withdrawal of HDACi. This mechanism is implemented as a consequence of the direct targeting of RAS by HDAC inhibition, which resulted in a further amplified GTP-binding activity and subsequent signalling through the MAPK pathway. The observed outcome was an increase in the priming of MYC for ubiquitin-mediated proteasomal degradation, thereby enabling the cells to exit the cell cycle and enter the protective state of G2 arrest. This process was functionally reversed with a conditional non-degradable MYC (T58A/S62A), which in turn rendered the cells more susceptible to undergo apoptosis. Conclusively, in the context of a constitutively activating RAS mutation, the prospect of HDACi treatment was effectively improved using current MAPK-targeted therapy by preventing the observed pro-oncogenic effect of the HDACi treatment alone. Note: This abstract was not presented at the meeting. Citation Format: Sylvia S. Ispasanie, Lena Boehme, Martin Eilers, Tilman Brummer, Kathleen Klotz-Noack, Natalia Kuhn, Bastian Gastl, Christine Sers. HDAC inhibitors and the mechanism of resistance in colorectal cancer: RAS and MYC - the partners in crime [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5170. doi:10.1158/1538-7445.AM2017-5170


Re-replication induced by geminin depletion occurs from G2 and is enhanced by checkpoint activation

February 2012

·

992 Reads

·

61 Citations

Journal of Cell Science

To prevent re-replication of DNA in a single cell cycle, the licensing of replication origins by Mcm2-7 is prevented during S and G2 phases. Animal cells achieve this by cell-cycle-regulated proteolysis of the essential licensing factor Cdt1 and inhibition of Cdt1 by geminin. Here we investigate the consequences of ablating geminin in synchronised human U2OS cells. Following geminin loss, cells complete an apparently normal S phase, but a proportion arrest at the G2-M boundary. When Cdt1 accumulates in these cells, DNA re-replicates, suggesting that the key role of geminin is to prevent re-licensing in G2. If cell cycle checkpoints are inhibited in cells lacking geminin, cells progress through mitosis and less re-replication occurs. Checkpoint kinases thereby amplify re-replication into an all-or-nothing response by delaying geminin-depleted cells in G2. Deep DNA sequencing revealed no preferential re-replication of specific genomic regions after geminin depletion. This is consistent with the observation that cells in G2 have lost their replication timing information. By contrast, when Cdt1 is overexpressed or is stabilised by the neddylation inhibitor MLN4924, re-replication can occur throughout S phase.


Citations (7)


... Next, we assessed differences in signaling pathway activities of colon cancer cell subpopulations with differential GPA33 mRNA expression, using single-cell transcriptome sequencing data of 12 samples of freshly resected colon cancers [16]. We grouped cancer cells into GPA33-high and -low clusters and looked for differences in pathway activities through assessment of expression of specific target gene signatures (Fig. 1F). ...

Reference:

GPA33 expression in colorectal cancer can be induced by WNT inhibition and targeted by cellular therapy
Mitogen‐activated protein kinase activity drives cell trajectories in colorectal cancer

EMBO Molecular Medicine

... In colorectal cancer, the mutation rate of KRAS reaches 40-50%, while the mutation rate of NRAS is approximately 5-9%. Studies have found that tumors with NRAS mutations exhibit resistance to EGFR treatment [12]. Therefore, NRAS can regulate the growth, migration, invasion, and other activities of tumor cells through multiple pathways, including the activation of PI3K/AKT and NF-κB signaling pathways [13]. ...

Mutation-specific effects of NRAS oncogenes in colorectal cancer cells
  • Citing Article
  • December 2020

Advances in Biological Regulation

... Insulin growth factor 1 receptor (IGF1R) is a tyrosine kinase receptor on the membrane, and can be targeted by drugs since it is overexpressed in tumor metastasis, drug resistance, and poor prognosis in cancers. MALAT1 binds to SFPQ releasing oncogene PTBP2 from SFPQ/PTBP2 complex in CRC [130] , while in the same cancers SFPQ depletion is synthetically lethal with BRAF V600E mutations [131] . SFPQ promotes lung cancer malignancy [131] , promotes the proliferation, migration and invasion in HCC [128] . ...

SFPQ Depletion Is Synthetically Lethal with BRAFV600E in Colorectal Cancer Cells

Cell Reports

... Although these oncogenic events produce DNA damage that facilitates tumorigenesis and tumor heterogeneity, they also create a reduction in MCM/CMG functional fidelity in tumor cells that is likely exploitable with CMG inhibitors (1). Consistent with these concepts, oncogenic K-Ras also causes fork stalling and RS (18,19), and cells expressing mutant K-Ras are selectively sensitive to the reduction of MCM licensing (20), suggesting that future MCM/ CMG inhibitors will synergize with K-Ras mutations to suppress tumor growth. In addition, inhibition of MCM/CMG reserves can selectively sensitize tumor cells (including those with mutant K-Ras) to fork-stalling chemotherapy drugs (11,12), suggesting that CMG inhibitors will have the potential to overcome chemo-resistance in the management of cancer. ...

Reduced replication origin licensing selectively kills KRAS-mutant colorectal cancer cells via mitotic catastrophe

Cell Death and Disease

... Additionally, combination of IC 50 doses of mTORC1/2, PI3K and MEK inhibitors were also effective in preclinical models of ovarian clear cell carcinoma [305]. Also, in KRAS-mutant pancreatic cancer, concurrent inhibition of RAF and ERK was similarly highly synergistic in inducing apoptotic cell death at low doses, whereas each blocker alone was only cytostatic [306]. This low-dose multidrug approach to decrease driver network signaling has also been proven to be an effective strategy to suppress cancer metastasis [307]. ...

Low-Dose Vertical Inhibition of the RAF-MEK-ERK Cascade Causes Apoptotic Death of KRAS Mutant Cancers

Cell Reports

... In metazoans, additional CDK-independent mechanisms control helicase loading and interfering with these mechanisms also causes over-replication [19][20][21][22] . For example, unscheduled helicase loading results in extensive DNA damage and loss of cellular viability in cultured human cells and Xenopus laevis egg extracts [23][24][25][26][27][28][29][30][31] . In Drosophila melanogaster, follicle cells undergo developmentally programmed over-replication at specific genomic loci and DSBs occur at sites of potential head-to-tail replication collisions [32][33][34][35] . ...

Re-replication induced by geminin depletion occurs from G2 and is enhanced by checkpoint activation

Journal of Cell Science

... Curiously, multiple Mcm2-7 complexes can be loaded in vitro and during initiation in cells, even though only two are destined to become part of an active replication fork (209,210). This so-called MCM paradox (211) differs from bacterial initiation, in which only two molecules of DnaB are deposited per round of origin firing (145,212) and may have a role in helping to overcome replication fork stalling, for example, by the activation of dormant origins (212). ...

A Role for Dormant Origins in Tumor Suppression
  • Citing Article
  • March 2011

Molecular Cell