Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marianne Koritzinsky is active.

Publication


Featured researches published by Marianne Koritzinsky.


Nature Reviews Cancer | 2008

Hypoxia signalling through mTOR and the unfolded protein response in cancer

Bradly G. Wouters; Marianne Koritzinsky

Hypoxia occurs in the majority of tumours, promoting angiogenesis, metastasis and resistance to therapy. Responses to hypoxia are orchestrated in part through activation of the hypoxia-inducible factor family of transcription factors (HIFs). Recently, two additional O2-sensitive signalling pathways have also been implicated: signalling through the mammalian target of rapamycin (mTOR) kinase and signalling through activation of the unfolded protein response (UPR). Although they are activated independently, growing evidence suggests that HIF-, mTOR- and UPR-dependent responses to hypoxia act in an integrated way, influencing each other and common downstream pathways that affect gene expression, metabolism, cell survival, tumorigenesis and tumour growth.


Molecular and Cellular Biology | 2002

Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha

Constantinos Koumenis; Christine Naczki; Marianne Koritzinsky; Sally Rastani; Alan J. Diehl; Nahum Sonenberg; Antonis E. Koromilas; Bradly G. Wouters

ABSTRACT Hypoxia profoundly influences tumor development and response to therapy. While progress has been made in identifying individual gene products whose synthesis is altered under hypoxia, little is known about the mechanism by which hypoxia induces a global downregulation of protein synthesis. A critical step in the regulation of protein synthesis in response to stress is the phosphorylation of translation initiation factor eIF2α on Ser51, which leads to inhibition of new protein synthesis. Here we report that exposure of human diploid fibroblasts and transformed cells to hypoxia led to phosphorylation of eIF2α, a modification that was readily reversed upon reoxygenation. Expression of a transdominant, nonphosphorylatable mutant allele of eIF2α attenuated the repression of protein synthesis under hypoxia. The endoplasmic reticulum (ER)-resident eIF2α kinase PERK was hyperphosphorylated upon hypoxic stress, and overexpression of wild-type PERK increased the levels of hypoxia-induced phosphorylation of eIF2α. Cells stably expressing a dominant-negative PERK allele and mouse embryonic fibroblasts with a homozygous deletion of PERK exhibited attenuated phosphorylation of eIF2α and reduced inhibition of protein synthesis in response to hypoxia. PERK−/− mouse embryo fibroblasts failed to phosphorylate eIF2α and exhibited lower survival after prolonged exposure to hypoxia than did wild-type fibroblasts. These results indicate that adaptation of cells to hypoxic stress requires activation of PERK and phosphorylation of eIF2α and suggest that the mechanism of hypoxia-induced translational attenuation may be linked to ER stress and the unfolded-protein response.


Journal of Clinical Investigation | 2010

The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5

Twan van den Beucken; Ludwig Dubois; Hanneke Niessen; Johan Bussink; Kim Savelkouls; Tom G. Keulers; Hilda Mujcic; Willy Landuyt; Jan Willem Voncken; Philippe Lambin; Albert J. van der Kogel; Marianne Koritzinsky; Bradly G. Wouters

Tumor hypoxia is a common microenvironmental factor that adversely influences tumor phenotype and treatment response. Cellular adaptation to hypoxia occurs through multiple mechanisms, including activation of the unfolded protein response (UPR). Recent reports have indicated that hypoxia activates a lysosomal degradation pathway known as autophagy, and here we show that the UPR enhances the capacity of hypoxic tumor cells to carry out autophagy, and that this promotes their survival. In several human cancer cell lines, hypoxia increased transcription of the essential autophagy genes microtubule-associated protein 1 light chain 3beta (MAP1LC3B) and autophagy-related gene 5 (ATG5) through the transcription factors ATF4 and CHOP, respectively, which are regulated by PKR-like ER kinase (PERK, also known as EIF2AK3). MAP1LC3B and ATG5 are not required for initiation of autophagy but mediate phagophore expansion and autophagosome formation. We observed that transcriptional induction of MAP1LC3B replenished MAP1LC3B protein that was turned over during extensive hypoxia-induced autophagy. Correspondingly, cells deficient in PERK signaling failed to transcriptionally induce MAP1LC3B and became rapidly depleted of MAP1LC3B protein during hypoxia. Consistent with these data, autophagy and MAP1LC3B induction occurred preferentially in hypoxic regions of human tumor xenografts. Furthermore, pharmacological inhibition of autophagy sensitized human tumor cells to hypoxia, reduced the fraction of viable hypoxic tumor cells, and sensitized xenografted human tumors to irradiation. Our data therefore demonstrate that the UPR is an important mediator of the hypoxic tumor microenvironment and that it contributes to resistance to treatment through its ability to facilitate autophagy.


Molecular and Cellular Biology | 2004

Hypoxia-Mediated Down-Regulation of Bid and Bax in Tumors Occurs via Hypoxia-Inducible Factor 1-Dependent and -Independent Mechanisms and Contributes to Drug Resistance

Janine T. Erler; Christopher Cawthorne; Kaye J. Williams; Marianne Koritzinsky; Bradley G Wouters; Claire Wilson; Crispin J. Miller; Costas Demonacos; Ian J. Stratford; Caroline Dive

ABSTRACT Solid tumors with disorganized, insufficient blood supply contain hypoxic cells that are resistant to radiotherapy and chemotherapy. Drug resistance, an obstacle to curative treatment of solid tumors, can occur via suppression of apoptosis, a process controlled by pro- and antiapoptotic members of the Bcl-2 protein family. Oxygen deprivation of human colon cancer cells in vitro provoked decreased mRNA and protein levels of proapoptotic Bid and Bad. Hypoxia-inducible factor 1 (HIF-1) was dispensable for the down-regulation of Bad but required for that of Bid, consistent with the binding of HIF-1α to a hypoxia-responsive element (positions −8484 to −8475) in the bid promoter. Oxygen deprivation resulted in proteosome-independent decreased expression of Bax in vitro, consistent with a reduction in global translation efficiency. The physiological relevance of Bid and Bax down-regulation was confirmed in tumors in vivo. Oxygen deprivation resulted in decreased drug-induced apoptosis and clonogenic resistance to agents with different mechanisms of action. The contribution of Bid and/or Bax down-regulation to drug responsiveness was demonstrated by the relative resistance of normoxic cells that had no or reduced expression of Bid and/or Bax and by the finding that forced expression of Bid in hypoxic cells resulted in increased sensitivity to the topoisomerase II inhibitor etoposide.


Cancer Biology & Therapy | 2006

Translational control of gene expression during hypoxia.

Twan van den Beucken; Marianne Koritzinsky; Bradly G. Wouters

Poor oxygenation is a unique and prevalent feature of solid tumors associated with poor patient prognosis. In part, this is caused by a series of adaptive cellular responses that together have a large impact on gene expression and cell phenotype. HIF plays a key role in this response by activating a transcriptional program that stimulates genes involved in angiogenesis, cell metabolism, cell survival and cell invasion. Recently, hypoxia has also been shown to suppress protein synthesis through the regulation of the initiation step of mRNA translation. This appears to be a common feature of the cell in response to hypoxia and is mediated by two distinct pathways. The first occurs rapidly, is transient, and is associated with activation of the unfolded protein response (UPR) that occurs in response to endoplasmic reticulum (ER) stress. Translation inhibition during this initial phase is due to phosphorylation of eukaryotic initiation factor 2α (eIF2α) in a PERK dependent manner. Although this effect is transient, overall levels of translation remain low during hypoxia due to inhibition of a second eukaryotic initiation complex, eIF4F. This second mechanism is multi-factorial, but due at least in part to inhibition of the mTOR kinase. Although each of these pathways leads to a general inhibition in translation, the consequence at the individual gene level is highly variable. This is due to sequences in the 5’ and 3’ untranslated regions (UTRs) of mRNA that confer their ability to maintain, or even increase, translation efficiency in spite of the overall inhibition. Consequently, regulation of mRNA translation appears to be an important mediator of gene expression during hypoxia.


Radiotherapy and Oncology | 2009

Autophagy is required during cycling hypoxia to lower production of reactive oxygen species

Chantal Ramaekers; Marco B.E. Schaaf; Tom G. Keulers; Kim Savelkouls; Philippe Lambin; Marianne Koritzinsky; Bradly G. Wouters

BACKGROUND AND PURPOSEnHuman tumors are characterized by the presence of cells that experience periodic episodes of hypoxia followed by reoxygenation. These cells are exposed to reactive oxygen species (ROS) upon reoxygenation and require adaptation to this stress by lowering ROS production or enhancing ROS-clearance for their survival. We hypothesized that autophagy, a lysosomal degradation pathway, may be involved in reducing ROS during periodic hypoxia through removal of ROS producing species.nnnMATERIALS AND METHODSnHuman tumor cells (MCF-7, HT29, U373) were exposed to cycles of hypoxia (O(2)<0.02%) and reoxygenation in the absence or presence of the autophagy inhibitor chloroquine (CQ). Clonogenic survival, ROS production and mitochondrial-DNA content were assessed. In addition, A549 cells overexpressing wild-type or K63-mutated ubiquitin (K63R) were analyzed for ROS production.nnnRESULTSnOur data indicate that CQ treatment sensitizes cells to cycling hypoxia, due to increased production of ROS, associated with an incapacity to reduce mitochondrial content. Addition of the ROS-scavenger N-acetyl-cysteine increased cell viability and neutralized CQ-effects. Additionally, genetic prevention of K63-linked ubiquitin chains that are required for the removal of toxic protein aggregates by autophagy, resulted in increased ROS production.nnnCONCLUSIONSnInhibition of autophagy substantially increases cell death induced by cycling hypoxia through increased ROS production, providing an opportunity to decrease the hypoxic fraction within tumors and enhance tumor therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2013

PERK/eIF2α signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS

Ludwig Dubois; Tom G. Keulers; Twan van den Beucken; Philippe Lambin; Johan Bussink; Albert J. van der Kogel; Marianne Koritzinsky; Bradly G. Wouters

Hypoxia is a common feature of tumors and an important contributor to malignancy and treatment resistance. The ability of tumor cells to survive hypoxic stress is mediated in part by hypoxia-inducible factor (HIF)-dependent transcriptional responses. More severe hypoxia activates endoplasmatic reticulum stress responses, including the double-stranded RNA-activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2α (eIF2α)-dependent arm of the unfolded protein response (UPR). Although several studies implicate important roles for HIF and UPR in adaption to hypoxia, their importance for hypoxic cells responsible for therapy resistance in tumors is unknown. By using isogenic models, we find that HIF and eIF2α signaling contribute to the survival of hypoxic cells in vitro and in vivo. However, the eIF2α-dependent arm of the UPR is uniquely required for the survival of a subset of hypoxic cells that determine tumor radioresistance. We demonstrate that eIF2α signaling induces uptake of cysteine, glutathione synthesis, and protection against reactive oxygen species produced during periods of cycling hypoxia. Together these data imply that eIF2α signaling is a critical contributor to the tolerance of therapy-resistant cells that arise as a consequence of transient changes in oxygenation in solid tumors and thus a therapeutic target in curative treatments for solid cancers.


Nature Communications | 2014

Hypoxia promotes stem cell phenotypes and poor prognosis through epigenetic regulation of DICER

Twan van den Beucken; Elizabeth Koch; Kenneth C. Chu; Rajesha Rupaimoole; Peggy Prickaerts; Michiel E. Adriaens; Jan Willem Voncken; Adrian L. Harris; Francesca M. Buffa; Syed Haider; Maud H. W. Starmans; Cindy Q. Yao; Mircea Ivan; Cristina Ivan; Chad V. Pecot; Paul C. Boutros; Anil K. Sood; Marianne Koritzinsky; Bradly G. Wouters

MicroRNAs are small regulatory RNAs that post-transcriptionally control gene expression. Reduced expression of DICER, the enzyme involved in microRNA processing, is frequently observed in cancer and is associated with poor clinical outcome in various malignancies. Yet the underlying mechanisms are not well understood. Here, we identify tumor hypoxia as a regulator of DICER expression in large cohorts of breast cancer patients. We show that DICER expression is suppressed by hypoxia through an epigenetic mechanism that involves inhibition of oxygen-dependent H3K27me3 demethylases KDM6A/B and results in silencing of the DICER promoter. Subsequently, reduced miRNA processing leads to derepression of the miR-200 target ZEB1, stimulates the epithelial to mesenchymal transition and ultimately results in the acquisition of stem cell phenotypes in human mammary epithelial cells. Our study uncovers a previously unknown relationship between oxygen-sensitive epigenetic regulators, miRNA biogenesis and tumor stem cell phenotypes that may underlie poor outcome in breast cancer.


Radiotherapy and Oncology | 2009

Hypoxic activation of the unfolded protein response (UPR) induces expression of the metastasis-associated gene LAMP3

Hilda Mujcic; Tomasz Rzymski; Marianne Koritzinsky; Manuela Milani; Adrian L. Harris; Bradly G. Wouters

BACKGROUND AND PURPOSEnTumour hypoxia contributes to failure of cancer treatment through its ability to protect against therapy and adversely influence tumour biology. In particular, several studies suggest that hypoxia promotes metastasis. Hypoxia-induced cellular changes are mediated by oxygen-sensitive signaling pathways that activate downstream transcription factors. We have investigated the induction and transcriptional regulation of a novel metastasis-associated gene, LAMP3 during hypoxia.nnnMATERIALS AND METHODSnMicroarray, quantitative PCR, Western blot analysis and immunohistochemistry were used to investigate hypoxic regulation of LAMP3. The mechanism for LAMP3 induction was investigated using transient RNAi and stable shRNA targeting components of the hypoxic response. Endoplasmic reticulum stress inducing agents, including proteasome inhibitors were assessed for their ability to regulate LAMP3.nnnRESULTSnLAMP3 is strongly induced by hypoxia at both the mRNA and protein levels in a large panel of human tumour cell lines. Induction of LAMP3 occurs as a consequence of the activation of the PERK/eIF2alpha/ATF4 arm of the unfolded protein response (UPR) and is independent of HIF-1alpha. LAMP3 is expressed heterogeneously within the microenvironment of tumours, overexpressed in breast cancer, and increases in tumours treated with avastin.nnnCONCLUSIONSnThese data identify LAMP3 as a novel hypoxia-inducible gene regulated by the UPR. LAMP3 is a new candidate biomarker of UPR activation by hypoxia in tumours and is a potential mediator of hypoxia-induced metastasis.


Journal of Biological Chemistry | 2009

Hypoxia-induced Expression of Carbonic Anhydrase 9 Is Dependent on the Unfolded Protein Response

Twan van den Beucken; Marianne Koritzinsky; Hanneke Niessen; Ludwig Dubois; Kim Savelkouls; Hilda Mujcic; Barry Jutten; Juraj Kopacek; Sylvia Pastorekova; Albert J. van der Kogel; Philippe Lambin; Willem Voncken; Bradly G. Wouters

Adaptation to tumor hypoxia is mediated in large part by changes in protein expression. These are driven by multiple pathways, including activation of the hypoxia inducible factor-1 (HIF-1) transcription factor and the PKR-like endoplasmic reticulum kinase PERK, a component of the unfolded protein response. Through gene expression profiling we discovered that induction of the HIF-1 target gene CA9 was defective in mouse embryo fibroblasts derived from mice harboring an eIF2α S51A knock-in mutation. This finding was confirmed in two isogenic human cell lines with an engineered defect in eIF2α phosphorylation. We show that impaired CA9 expression was not due to changes in HIF activity or CA9 mRNA stability. Using chromatin immunoprecipitation we show that the eIF2α-dependent translationally regulated gene ATF4 binds directly to the CA9 promoter and is associated with loss of the transcriptional repressive histone 3 lysine 27 tri-methylation mark. Loss or overexpression of ATF4 confirmed its role in CA9 induction during hypoxia. Our data indicate that expression of CA9 is regulated through both the HIF-1 and unfolded protein response hypoxia response pathways in vitro and in vivo.

Collaboration


Dive into the Marianne Koritzinsky's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philippe Lambin

Maastricht University Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Ravi N. Vellanki

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Dan Cojocari

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Anthony M. Joshua

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Ludwig Dubois

Maastricht University Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Hilda Mujcic

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Albert J. van der Kogel

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge