Jukka Westermarck’s research while affiliated with Turku centre for biotechnology, finland 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 (227)


Germline-specific RNA helicase DDX4 forms cytoplasmic granules in cancer cells and promotes tumor growth
  • Article
  • Full-text available

July 2024

·

96 Reads

Cell Reports

·

·

·

[...]

·

Cancer cells undergo major epigenetic alterations and transcriptomic changes, including ectopic expression of tissue- and cell-type-specific genes. Here, we show that the germline-specific RNA helicase DDX4 forms germ-granule-like cytoplasmic ribonucleoprotein granules in various human tumors, but not in cultured cancer cells. These cancerous DDX4 complexes contain RNA-binding proteins and splicing regulators, including many known germ granule components. The deletion of DDX4 in cancer cells induces transcriptomic changes and affects the alternative splicing landscape of a number of genes involved in cancer growth and invasiveness, leading to compromised capability of DDX4-null cancer cells to form xenograft tumors in immunocompromised mice. Importantly, the occurrence of DDX4 granules is associated with poor survival in patients with head and neck squamous cell carcinoma and higher histological grade of prostate cancer. Taken together, these results show that the germ-granule-resembling cancerous DDX4 granules control gene expression and promote malignant and invasive properties of cancer cells.

Download

DUSP6 inhibition overcomes neuregulin/HER3-driven therapy tolerance in HER2+ breast cancer

June 2024

·

14 Reads

·

6 Citations

EMBO Molecular Medicine

Despite clinical benefits of tyrosine kinase inhibitors (TKIs) in cancer, most tumors can reactivate proliferation under TKI therapy. Here we present transcriptional profiling of HER2+ breast cancer cells transitioning from dormant drug tolerant cells to re-proliferating cells under continuous HER2 inhibitor (HER2i) therapy. Focusing on phosphatases, expression of dual-specificity phosphatase DUSP6 was found inhibited in dormant cells, but strongly induced upon regrowth. DUSP6 expression also selectively associated with poor patient survival in HER2+ breast cancers. DUSP6 overexpression conferred apoptosis resistance, whereas its pharmacological blockade prevented therapy tolerance development under HER2i therapy. DUSP6 targeting also synergized with clinically used HER2i combination therapies. Mechanistically DUSP6 is a positive regulator of HER3 expression, and its impact on HER2i tolerance was mediated by neuregulin-HER3 axis. In vivo, genetic targeting of DUSP6 reduced tumor growth in brain metastasis model, whereas its pharmacological targeting induced synthetic lethal therapeutic effect in combination with HER2i. Collectively this work demonstrates that DUSP6 drives escape from HER2i-induced dormancy, and that DUSP6 is a druggable target to overcome HER3-driven TKI resistance.


Abstract 1717: RAS and PP2A co-regulated phosphosite on KDM1A dictates its substrate specificity & transcriptional outcome

March 2024

·

2 Reads

Cancer Research

RAS-mediated human cell transformation requires simultaneous inhibition of tumor suppressor phosphatase PP2A; however, the target phosphosites co-regulated by RAS and PP2A are poorly characterized. In this study, we describe the phosphoregulation of an epigenetic regulator, KDM1A, by RAS and PP2A. KDM1A is a lysine-specific demethylase that can act as a transcriptional activator or inhibitor, depending on its specific activity toward various histone marks. However, the post-translational modifications regulating the substrate specificity of KDM1A are poorly addressed. We found that KDM1A physically interacts with PP2A through a conserved binding motif, and that mutating the motif residues decreased its interaction with PP2A. Additionally, PP2A activation or RAS inhibition led to increased chromatin recruitment of KDM1A. This enhanced chromatin recruitment of KDM1A was recapitulated upon CRISPR/CAS9-mediated substitution of the PP2A/RAS-targeted serine to alanine. Using histone proteomics, we further identified that the RAS/PP2A-regulated phosphosite determines the substrate specificity of KDM1A. This is evidenced by an increased demethylation of histone H3K9me2/3, a marker of gene activation. The differential gene expression profile of the phosphorylation mutants of KDM1A predominantly showed increased gene expression, further validating our findings. The gene expression profiles of KDM1A phosphomutant RAS mutant cells demonstrate gene level specificity indicating that this newly discovered KDM1A phosphoswitch has an important role in specifying KDM1A function in cancer. Collectively, our results demonstrate that RAS and PP2A activities converge on phosphoregulation of epigenetic machinery in cancer cells. This RAS and PP2A co-regulated phosphosite on KDM1A dictates both its histone mark specificity and transcriptional outcome. Citation Format: Mukund Sharma, Anna Aakula, Jesse Kamila, Reetta Nätkin, Tiziana Bonaldi, Saverio Minucci, Matti Nykter, Jukka Westermarck. RAS and PP2A co-regulated phosphosite on KDM1A dictates its substrate specificity & transcriptional outcome [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 1717.



PP2A-based triple-strike therapy overcomes mitochondrial apoptosis resistance in brain cancer cells

July 2023

·

46 Reads

·

5 Citations

Mitochondrial glycolysis and hyperactivity of the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) pathway are hallmarks of malignant brain tumors. However, kinase inhibitors targeting AKT (AKTi) or the glycolysis master regulator pyruvate dehydrogenase kinase (PDKi) have failed to provide clinical benefits for brain tumor patients. Here, we demonstrate that heterogenous glioblastoma (GB) and medulloblastoma (MB) cell lines display only cytostatic responses to combined AKT and PDK targeting. Biochemically, the combined AKT and PDK inhibition resulted in the shutdown of both target pathways and priming to mitochondrial apoptosis but failed to induce apoptosis. In contrast, all tested brain tumor cell models were sensitive to a triplet therapy, in which AKT and PDK inhibition was combined with the pharmacological reactivation of protein phosphatase 2A (PP2A) by NZ-8-061 (also known as DT-061), DBK-1154 and DBK-1160. We also provide proof-of-principle evidence for in vivo efficacy in the intracranial GB and MB models by the brain-penetrant triplet therapy (AKTi + PDKi + PP2A reactivator). Mechanistically, PP2A reactivation converted the cytostatic AKTi + PDKi response to cytotoxic apoptosis, through PP2A-elicited shutdown of compensatory mitochondrial oxidative phosphorylation and by increased proton leakage. These results encourage the development of triple-strike strategies targeting mitochondrial metabolism to overcome therapy tolerance in brain tumors.


Figure 2: Clinical association of DUSP6 overexpression with poor prognosis HER2+ breast
Figure 3: Functional involvement of DUSP6 in HER2i tolerance development.
Figure 4: Both genetic and pharmacological DUSP6 targeting overcomes HER2i resistance
Figure 5: DUSP6 inhibition overcomes HER2 inhibitor resistance in vivo. (A) DUSP6 knockout
Fig. 7. Identification of HER3-DUSP6 positive feedback loop. (A) DUSP6 knockdown inhibits HER2
Transcriptional landscape of DTP-DTEP transition reveals DUSP6 as a driver of HER2 inhibitor tolerance via Neuregulin/HER3 axis

June 2023

·

98 Reads

The mechanisms promoting re-growth of dormant cancer cells under continuous tyrosine kinase inhibitor (TKI) therapy are poorly understood. Here we present transcriptional profiling of HER2+ breast cancer cells treated continuously with HER2 TKI (HER2i) therapy for 9 months. The data reveals specific gene regulatory programs associated with transition from dormant drug tolerant persister cells (DTPs) to proliferating DTEP (drug tolerant expanding persister) cells and eventually long-term resistance. Focusing on yet poorly understood phosphatases as determinants of therapy tolerance, expression of dual-specificity phosphatase DUSP6 was found inhibited in DTPs, but strongly induced upon re-growth of DTEPs. DUSP6 overexpression conferred apoptosis resistance whereas its pharmacological blockade prevented DTEP development under HER2i therapy. The DUSP6-driven HER2i tolerance was mediated by activation of neuregulin-HER3 axis, and consistent with the role of HER3 in widespread therapy tolerance, DUSP6 targeting also synergized with clinically used HER2i combination therapies. In vivo, pharmacological DUSP6 targeting induced synthetic lethal effect with HER2i in independent tumor models, and its genetic targeting reduced tumor growth in orthotopic brain metastasis model. Collectively this work provides first transcriptional landscape of DTP-DTEP transition under TKI therapy, and identify DUSP6 as a novel candidate therapy target to overcome widespread HER3-driven therapy resistance.


Figure 3. ROS induction is required for tumor cell death in LG conditions (A) Analysis of ROS levels measured in HeLa and HCT116 cells treated for 36 h with vehicle, IACS-010759, or antimycin A in either HG (10 mM) or LG (2.5 mM) conditions, measured by cellROX (n = 2) or amplexRED (n = 16) assays. (B) Proliferation and survival data of HeLa and HCT116 cells treated with 0.5 mM H 2 O 2 for 24 h, cultured either in standard conditions (10 mM glucose: HG) or in glucose restriction (2.5 mM glucose: LG). Left panels show normalized count of cells, right panels quantification of death by propidium iodide exclusion assay. Data refer to two independent experiments with three replicates each experimental point (n = 6, graph shows mean ± SD). (C) Proliferation and survival data of HeLa and HCT116 cells cultured 48 h following the protocol described in Figure 1. NAC (5 mM added every 8 h) was added as indicated. Left panels show normalized count of cells, right panels quantification of death by propidium iodide exclusion assay. Data refer to two independent experiments with three replicates each experimental point (n = 6, graph shows mean ± SD).
Figure 4. ROS mediate CIP2A downregulation and downstream activation of the PP2A-GSL3b-MCL1 signaling pathway (A) Immunoblotting analysis to measure CIP2A levels in cell lysates from HeLa and HCT116 cells treated with 0.5mM H 2 O 2 for 24 h, cultured in standard conditions. Tubulin was used as a loading control. (B) Immunoblotting analysis to measure CIP2A levels in cell lysates from HeLa and HCT116 cells treated with IACS-010759 for 48 h, cultured in standard conditions. NAC (5 mM added every 8 h) and chloroquine (20 mM) were added as indicated. Vinculin was used as a loading control. (C) Immunoblotting analysis to measure levels of pGSK3b or GSK3b, and of MCL1. Vinculin was used as a loading control. HeLa and HCT116 cells were cultured either in standard conditions (10 mM glucose: HG) or in glucose restriction (2.5 mM glucose: LG). To mimic the in vivo ad libitum feeding and/or intermittent fasting cycles in an in vitro setting, cells were treated for 24 h in standard conditions, then media were replenished either with standard conditions (HG) or glucose starvation (LG) for other 24 h. NAC (5 mM added every 8 h) was added as indicated.
Figure 5. CIP2A degradation, by cooperative action of CMA and the proteasome, mediates the induction of cell death by IACS-010759 in LG conditions (A) Immunoblotting analysis to measure levels of the indicated proteins, derived from HeLa or HCT116 cells treated as indicated, transduced to express either scrambled shRNA as a control (SCR) or an shRNA targeting LAMP2A. Tubulin was used as a loading control. (B) Immunoblotting analysis of CIP2A levels from cell lysates derived from HeLa and HCT116 cells treated with 0.5 mM H 2 O 2 for 24 h, transduced to express either scrambled shRNA as a control (SCR) or an shRNA targeting LAMP2A. (C) Proliferation and survival data of HeLa and HCT116 cells, transduced to express either scrambled shRNA as a control (SCR) or shRNAs targeting LAMP2A. Cells were cultured either in standard conditions (10 mM glucose: HG) or in glucose restriction (2.5 mM glucose: LG). To mimic the in vivo ad libitum feeding and/or intermittent fasting cycles in an in vitro setting, cells were treated for 24 h in standard conditions, then media were replenished either with standard conditions (HG) or glucose starvation (LG) for other 24 h. Left panels show normalized count of cells, right panels quantification of death by propidium iodide exclusion assay. Data refer to two independent experiments with three replicates each experimental point (n = 6, graph shows graph shows mean ± SD).
Figure 6. IACS-010759 triggers the ROSdependent dissociation of CIP2A from PP2A Proximity ligation assays (PLAs) were performed on LAMP2A knockdown HeLa cells. Cells were either treated with vehicle (DMSO), or treated with IACS-010759 90 nM, or treated with IACS-010759 90 nM plus NAC (5 mM added every 8 h). Left panel shows the quantification of PLA dots (1 dot = 1 interaction between CIP2A and PP2A subunit C). ****p < 0.005; ns, non-significant. Panels on the right show representative images of DAPI-stained nuclei, PLA reactions, and merged signals. Bottom row (only PLA probes) shows a control with PLA probes only. Data are representative of two independent experiments with 10 acquisitions over each experimental point (n = 20, mean and values). Scale bar, 10 mm.
Endogenous PP2A inhibitor CIP2A degradation by chaperone-mediated autophagy contributes to the antitumor effect of mitochondrial complex I inhibition

June 2023

·

127 Reads

·

9 Citations

Cell Reports

Combined inhibition of oxidative phosphorylation (OXPHOS) and glycolysis has been shown to activate a PP2A-dependent signaling pathway, leading to tumor cell death. Here, we analyze highly selective mitochondrial complex I or III inhibitors in vitro and in vivo to elucidate the molecular mechanisms leading to cell death following OXPHOS inhibition. We show that IACS-010759 treatment (complex I inhibitor) induces a reactive oxygen species (ROS)-dependent dissociation of CIP2A from PP2A, leading to its destabilization and degradation through chaperone-mediated autophagy. Mitochondrial complex III inhibition has analogous effects. We establish that activation of the PP2A holoenzyme containing B56δ regulatory subunit selectively mediates tumor cell death, while the arrest in proliferation that is observed upon IACS-010759 treatment does not depend on the PP2A-B56δ complex. These studies provide a molecular characterization of the events subsequent to the alteration of critical bioenergetic pathways and help to refine clinical studies aimed to exploit metabolic vulnerabilities of tumor cells.


PME‐1 overexpression associates with PTEN loss and therapy relapse of PTEN‐negative prostate cancer patients. (A) PME‐1 protein expression in PCa tissue microarray material from 358 patients treated primarily with radical prostatectomy in the Helsinki University Hospital between 1983 and 1998 was assessed by immunohistochemical stainings. PME‐1 protein content was scored using 4‐tier scale; negative (not shown), low, intermediate and strong expression. In subsequent clinical analysis negative‐intermediate immunopositivity of PME‐1 was considered as PME‐1 Low, whereas strong PME‐1 immunopositivity was considered as PME‐1 High. Scale bar 1 mm. (B) Kaplan‐Meyer analysis of time to secondary therapies after primary treatment based on PME‐1 status alone. P‐value by log rank test. (C) Assessment of the status of PME‐1 and PTEN in the patient populations and the putative effect on PP2A activity. (D, E) Kaplan‐Meyer analysis of time to secondary therapies after primary treatment based on PME‐1 status in combination with PTEN deletion (D) and AR expression (E). P‐values by log rank test.
PME‐1 promotes anchorage‐independent growth of prostate cancer cells. (A) The effect of PME‐1 depletion, using two independent siRNAs, was investigated by colony formation assays (10 days of growth) in two PTEN‐deficient PCa cells lines PC‐3 and DU‐145. (Left) Representative images of the wells. (Right) Bar graphs depicting the quantified data, mean ± SD of one (PC‐3) or two (DU‐145) independent experiments. (B) The effect of PME‐1 knock‐down on anchorage‐independent growth in soft agar assays (14 days of growth) in both PC‐3 and DU‐145 cells. (Left) Representative images depicting the colonies. (Right) Bar graphs displaying the quantified data, mean ± SD of two independent experiments. *P < 0.05, Welch's t‐test.
PME‐1 promotes anoikis resistance in PTEN‐deficient prostate cancer cells. (A) siCtrl‐ and siPME‐1‐transfected PC‐3 cells were plated 72 h post‐transfection on normal cell culture or low‐attachment plates for 24 h, before collection and lysis, and subsequently analyzed by western blotting for cleaved PARP‐1 (cPARP (p25)). Shown is a representative result from six independent experiments. (B) Apoptosis induction, as measured by PARP cleavage, in CRISPR/Cas9 generated PC‐3 cells, with and without non‐targeting gRNA (lanes 1–3), a pool of PME‐1 gRNA transfected cells (lane 4), and a single cell subclone of PME‐1 targeted cells (lane 5), after 24 h detachment. Shown is a representative result from three independent experiments. (C) The effect of stable PME‐1 overexpression in mouse embryonic fibroblasts (MEFs) from a PTEN/p53KO mouse model, after 24 h of detachment, was assessed by western blotting for cleaved PARP. Shown is a representative result from two independent experiments. (D) Mechanosensitive PARP cleavage in PME‐1‐depleted PC‐3 cells. The cells were cultured on soft (0.5 kPa) or stiff (50 kPa) polyacrylamide hydrogel for 24 h before being scraped into PBS, spun down, lyzed and analyzed for protein expression. Shown is a representative result from four independent experiments. (E) The number of Annexin V positive objects, a marker for apoptosis, in cultures of siCtrl‐ and siPME‐1‐transfected PC‐3 cells upon varying degrees of cell‐matrix interaction. The cells were grown on PEG (Poly(ethylene glycol))‐coated surfaces with 0–20% biotinylated compound bound to integrin ligand, fibronectin fragment FNIII(7–10). STS = staurosporine. Mean ± SD of three independent experiments. (F) Annexin V positive objects in PC‐3 cultures with no or high levels of integrin ligand, after 60 h of incubation. Mean ± SD of three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001, one‐way ANOVA with Bonferroni's multiple‐comparison test.
PME‐1 supports in vivo anoikis resistance and survival of prostate cancer cells in circulation. (A) The effect of PME‐1 on anchorage‐independent growth of PC‐3 cells on chick chorioallantoic membrane (CAM). PC‐3 cells were transiently transfected with either control siRNA (siCtrl) or PME‐1‐targeting siRNA (siPME‐1), and 24 h post‐transfection placed on the CAM. Growth of tumours was followed for 3–5 days. Shown are representative examples from three replicate experiments. (B) Immunohistological staining of dissected tumours using antibodies for PME‐1, Vimentin, TUNEL and Ki67. Shown are representative images from three (Ki67) or two (TUNEL) replicate experiments. Scale bar 50 μm for PME‐1 and Vimentin stainings and 100 μm for TUNEL and Ki67. (C) Quantification of TUNEL‐ and Ki‐67‐positive nuclei in the excised CAM tumours. Mean ± SD from a representative experiment with four eggs per condition, *P < 0.05, Mann–Whitney test. (D) The survival of siCtrl‐ and siPME‐1‐transfected PC‐3 cells in circulation. The cells were microinjected into the common cardinal veins of zebrafish embryos 72 h post‐transfection. After overnight incubation the embryos were imaged by fluorescence stereomicroscopy. Scale bar 500 μm. (E) The number of surviving fluorescent tumour cells per embryo from (D). Box plots depicting the range, 25th, 50th and 75th percentiles of the data overlaid with the individual data points combined from three independent experiments. **P < 0.01, Mann–Whitney test.
The potential mechanisms for PME‐1‐mediated anoikis resistance. (A) Immunofluorescence images (left) and quantification (right) depicting phosphorylated FAK (Y397) and early endosomal marker EEA1 in control and PME‐1‐depleted PC‐3 cells grown on soft (0.5 kPa) hydrogels for 24 h. Scale bar, 10 μm (main), 2 μm (inset). Mean ± SD from a representative of two independent experiments. (B) Fluorescence images depicting filamentous actin in control and PME‐1 KO PC‐3 cells treated with DMSO or increasing concentrations of Cytochalasin D (0.1–10 μm). Scale bar, 20 μm. Shown is a representative of two independent experiments. (C) Western blot (top) and quantification (bottom) displaying PARP cleavage in PC‐3 cells upon actin cytoskeleton disruption with Cytochalasin D. Mean ± SD of two independent experiments. (D, E) Images (D) and quantification (E) showing nuclear deformation in PC‐3 cells subjected to micropipette aspiration, as described in [46], over the course of 2 min. Mean ± SD of three independent experiments. *P < 0.05, Welch's t‐test. (F, G) Quantification of GFP‐NLS‐positive cells with cytoplasmic GFP signal (F) and the percentage of transfected cells with visible cGAS‐mCherry foci (G), respectively, in siCtrl‐ and siPME‐1‐treated cells grown on soft (0.5 kPa) substrate. Both readouts serve as a surrogate measure of compromised nuclear envelope. Mean ± SD of two independent experiments. **P < 0.01, Welch's t‐test (H–I). Immunofluorescence images and quantification depicting H3K9me3 (H) and H3K27me3 (I) levels in transiently PME‐1‐depleted and control PC‐3 cells, 48 h post‐transfection and 24 h after seeding on soft (0.5 kPa) substrate. Scale bar, 10 μm. Mean ± SD, representative of two independent experiments. ***P < 0.001, Mann–Whitney test. (J) Schematic representation of a putative model for including PME‐1 and its role in anoikis sensitivity/resistance in PCa diagnostics and therapy decision.
PP2A methylesterase PME-1 suppresses anoikis and is associated with therapy relapse of PTEN-deficient prostate cancers

April 2023

·

94 Reads

·

10 Citations

While organ-confined prostate cancer (PCa) is mostly therapeutically manageable, metastatic progression of PCa remains an unmet clinical challenge. Resistance to anoikis, a form of cell death initiated by cell detachment from the surrounding extracellular matrix, is one of the cellular processes critical for PCa progression towards aggressive disease. Therefore, further understanding of anoikis regulation in PCa might provide therapeutic opportunities. Here, we discover that PCa tumors with concomitant inhibition of two tumor suppressor phosphatases, PP2A and PTEN, are particularly aggressive, having less than 50% 5-year secondary-therapy-free patient survival. Functionally, overexpression of PME-1, a methylesterase for the catalytic PP2A-C subunit, inhibits anoikis in PTEN-deficient PCa cells. In vivo, PME-1 inhibition increased apoptosis in in ovo PCa tumor xenografts, and attenuated PCa cell survival in zebrafish circulation. Molecularly, PME-1-deficient PC3 cells display increased trimethylation at lysines 9 and 27 of histone H3 (H3K9me3 and H3K27me3), a phenotype known to correlate with increased apoptosis sensitivity. In summary, our results demonstrate that PME-1 supports anoikis resistance in PTEN-deficient PCa cells. Clinically, these results identify PME-1 as a candidate biomarker for a subset of particularly aggressive PTEN-deficient PCa.


Abstract 4727: RAS and PP2A activities converge on phosphoregulation of epigenetic complexes in cancer

April 2023

·

12 Reads

Cancer Research

RAS-mediated human cell transformation requires inhibition of the tumor suppressor Protein Phosphatase 2A (PP2A). Both RAS and PP2A mediate their effects by phosphoregulation, but phosphoprotein targets in which RAS and PP2A activities converge in human cancers have not been systematically analyzed. Here, based on mass spectrometry phosphoproteome data, we discover that phosphosites co-regulated by RAS and PP2A are enriched on proteins involved in epigenetic gene regulation. As examples, RAS and PP2A co-regulate the same phosphorylation sites on HDAC1/2, KDM1A, MTA1/2, RNF168 and TP53BP1. Mechanistically, we validate co-regulation of NuRD chromatin repressor complex by RAS and PP2A. Consistent with their known synergistic effects in cancer, RAS activation and PP2A inhibition resulted in epigenetic reporter de-repression and activation of oncogenic transcription. Notably, transcriptional de-repression by PP2A inhibition was associated with increased euchromatin and decrease in global DNA methylation. In RAS-driven cancers, PP2A activity is inhibited by nuclear PP2A inhibitor proteins PME-1 and SET. We further demonstrate that these PP2A inhibitors drive oncogenic transcription, but that their transcriptional targets are highly diversified. Whereas PME-1 controls RAS and MYC-driven transcription, SET controls TP53 targets and G2/M checkpoint genes. We further provide evidence that these nuclear PP2A inhibitors significantly differ in their roles in regulating DNA methylation. Collectively the results indicate that epigenetic protein complexes involved in oncogenic gene expression constitute a significant point of convergence for RAS hyperactivity and PP2A inhibition in cancer. Further, the results provide a rich source for future understanding of phosphorylation as a previously unappreciated layer of regulation of epigenetic gene regulation in cancer, and in other RAS/PP2A-regulated cellular processes. Citation Format: Mukund Sharma, Anna Aakula, Francesco Tabaro, Henrik Honkanen, Jesse Kamila, Matthieu Schapira, Cheryl Arrowsmith, Matti Nykter, Jukka K. Westermarck. RAS and PP2A activities converge on phosphoregulation of epigenetic complexes in cancer. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4727.


Triplet mitochondrial therapy. The reviewed evidence reveals the therapeutic potential of targeting cancer cells with triplet therapy combinations including one component from each corner of the triangle. Importantly, many current cancer therapies also impact mitochondrial metabolism and/or membrane potential and therefore could be considered as mitochondrial targeting approaches. AKT, AKT serine/threonine kinase 1; BH3, BH3‐only protein; PI3K, phosphoinositol 3 kinase; PP2A, protein phosphatase 2A.
Inhibition of adaptive therapy tolerance in cancer: is triplet mitochondrial targeting the key?

March 2023

·

22 Reads

·

8 Citations

Targeted therapies have become a mainstay in the treatment of cancer, but their long-term efficacy is compromised by acquired drug resistance. Acquired therapy resistance develops via two phases - first through adaptive development of non-genetic drug tolerance which is followed by stable resistance through acquisition of genetic mutations. Drug tolerance has been described in practically all clinical cancer treatment contexts, and detectable drug-tolerant tumors are highly associated with treatment relapse and poor survival. Thereby novel therapeutic strategies are needed to overcome cancer therapy tolerance. Recent studies have identified a critical role of mitochondrial mechanisms in defining cancer cell sensitivity to targeted therapies and the surprising effects of established cancer therapies on mitochondria. Here, these recent studies are reviewed emphasizing an emerging concept of triplet therapies including three compounds targeting different cancer cell vulnerabilities, but including at least one compound that targets the mitochondria. These mitochondria-targeting triplet therapies have very promising preclinical effects in overcoming cancer therapy tolerance. Potential strategies of how to overcome challenges in clinical translation of mitochondria targeting triplet therapies are also discussed.


Citations (55)


... These procedures followed previous descriptions [19]. Quantitative reverse transcription-PCR (qRT-PCR) analysis was conducted on a QuantStudio Real-Time PCR instrument (ThermoFisher, Houston, TX, USA) using PowerUp SYBR Green Master Mix (ThermoFisher, Houston, TX, USA). ...

Reference:

SSTR2-Targeted Theranostics in Hepatocellular Carcinoma
DUSP6 inhibition overcomes neuregulin/HER3-driven therapy tolerance in HER2+ breast cancer
  • Citing Article
  • June 2024

EMBO Molecular Medicine

... 29,30 CIP2A is a functional oncogene that inhibits the PP2A complex during the DNA damage response, mitotic entry, and mitotic progression by replacing the structural subunit of the PP2A complex, blocking phosphatase function. 31,32 This co-dependency suggests that cell lines do not tolerate the loss of multiple negative regulators of PP2A activity. ...

CIP2A coordinates phosphosignaling, mitosis, and the DNA damage response
  • Citing Article
  • October 2023

Trends in Cancer

... PP2A is suggested to be a tumor suppressor across various cancers (7)(8)(9). The activity of PP2A is inhibited in human malignancies, including brain cancer, promoting malignant transformation of normal cells (10)(11)(12)(13). PP2A inhibition by either genetic mechanisms or overexpression of its endogenous inhibitor proteins, inhibitor 2 of protein phosphatase 2A (I2PP2A, SET) or cancerous inhibitor of protein phosphatase 2A (CIP2A), promotes tumorigenesis and correlates with therapeutic resistance (14)(15)(16). ...

PP2A-based triple-strike therapy overcomes mitochondrial apoptosis resistance in brain cancer cells
  • Citing Article
  • July 2023

... Currently, various drugs are used as OXPHOS inhibitors, mostly targeting respiratory complexes I, II, and III (CI-III), ATP synthase, disrupting MMP, or blocking specific TCA cycle enzymes (Carter et al. 2020, Cazzoli et al. 2023, Zeng & Hu 2023, Lu et al. 2024, Udumula et al. 2024. One of the first drugs in the class of mitochondrial complex inhibitors is IACS-010759 that selectively inhibits CI activity and impairs OXPHOS (Cazzoli et al. 2023). ...

Endogenous PP2A inhibitor CIP2A degradation by chaperone-mediated autophagy contributes to the antitumor effect of mitochondrial complex I inhibition

Cell Reports

... Taking the nearest associated gene, these areas of accessibility were associated with genes enriched in pathways such as epithelial to mesenchymal transition (EMT), inflammatory response, mitotic spindle, KRAS signaling up, and TGF signaling (Fig. 5E), consistent with our RNAseq results. Similar pathways were identified in response to global PP2A suppression in HeLa cells, indicating that these may be a part of core PP2A-regulated pathways [28]. Furthermore, the enriched motifs identified using de novo motif discovery algorithm, HOMER, were associated with developmental transcription factors (Prdm1, Hoxc13, Nkx2-3), hematopoietic transcription factors (Tcf1, Klf1) downstream regulators of TGF-β signaling (Smad3, Smad5), and inflammation (Stat3, Irf4, Irf3, Irf7) (Fig. 5F). ...

RAS and PP2A activities converge on epigenetic gene regulation

Life Science Alliance

... 29,30 CIP2A is a functional oncogene that inhibits the PP2A complex during the DNA damage response, mitotic entry, and mitotic progression by replacing the structural subunit of the PP2A complex, blocking phosphatase function. 31,32 This co-dependency suggests that cell lines do not tolerate the loss of multiple negative regulators of PP2A activity. ...

Structural mechanism for inhibition of PP2A-B56α and oncogenicity by CIP2A

... This double inhibition is considered an interesting strategy for treating cancer and other proliferative diseases, as it enhances the cytotoxicity of conventional chemotherapeutic drugs. This contributes to demonstrating that the Warburg effect is not solely based on high LDHA expression, as both isozymes need to be targeted to avoid fermentative glycolysis [25,88,89]. However, there are some hybrids (15b, 16c, 17c, and 18a) which are located differently in the enzyme compared to W31 (Figure 8). ...

Inhibition of adaptive therapy tolerance in cancer: is triplet mitochondrial targeting the key?

... Increased PME-1 expression and decreased LCMT-1 expression associated with decreased PP2Ac methylation levels were observed in the brains of AD patients [15,16]. Furthermore, the increase in PME-1 expression and reduced levels of carboxymethyl PP2Ac are detected in multiple cancer types and correlate with a worse prognosis of gliomas, liver cancer, and prostate cancer [17][18][19]. ...

PP2A methylesterase PME-1 suppresses anoikis and is associated with therapy relapse of PTEN-deficient prostate cancers

... cell proliferation regulating inhibitor of protein phosphatase 2a (ciP2a) is a dysregulated protein in several types of cancer, including breast cancer, colorectal cancer, bladder cancer and hepatocellular carcinoma, which can affect cell proliferation, cell cycle progression, apoptosis and tumor formation (15)(16)(17). in addition, ciP2a serves a critical role in nerve diseases, such as inhibiting depression-like behaviors and promoting the development of alzheimer's disease (18)(19)(20). The expression of eMT markers, including Snail, Vimentin and e-cadherin, has been shown to be regulated by ciP2a (21,22). ...

CIP2A deficiency promotes depression-like behaviors in mice through inhibition of dendritic arborization

EMBO Reports

... 171 Protein phosphorylation extensively regulates cancer cell proliferation, metastasis, and invasion. 172 For example, DAPK3 directly phosphorylates Ser556 of ULK1, which increases the activity of ULK1 and promotes the formation of the ULK1 complex, leading to inhibition of the proliferation of gastric cancer cells. The downregulation of DAPK3 in gastric cancer patients is related to poor prognosis. ...

SHARPIN serine 146 phosphorylation mediates ARP2/3 interaction, cancer cell invasion, and metastasis

Journal of Cell Science