Therapeutic Targeting of the Unfolded Protein Response in Cancer

A special issue of Cancers (ISSN 2072-6694).

Deadline for manuscript submissions: closed (31 December 2019) | Viewed by 39869

Special Issue Editor

INSERM U1242, University of Rennes, 35042 Rennes, France
Interests: endoplasmic reticulum; proteostasis; unfolded protein response; cancer; PERK; IRE1; ATF6; therapeutic approach

Special Issue Information

Dear Colleagues,

In recent years, endoplasmic reticulum (ER) proteostasis has been identified as a major player in cancer. In particular, the signaling mechanisms that are involved in ER proteostasis maintenance were shown to significantly contribute to cancer hallmarks such as epithelial-to-mesenchymal transition (EMT), inflammation, migration/invasion, or resistance to therapy. These signaling mechanisms are collectively assembled under the name of unfolded protein response (UPR) and transduced by three ER resident proteins named IRE1, PERK, and ATF6. The role of IRE1 and PERK in cancer development and their relevance as therapeutic targets in the disease have clearly been documented, whereas the role of ATF6 in oncogenesis or treatment resistance remains less clear.

In the past five years, a large number of pharmacological or genetic approaches targeting the three arms of the UPR were developed to be applied as therapeutics in various diseases, including cancer.

In this Special Issue, we aim at providing (i) state-of-the-art reviews on the current knowledge regarding the pharmacological and genetic tools that were developed to selectively target ATF6, PERK, IRE1 or UPR regulators, as well as their reported utilizations in cancer models; (ii) research articles describing novel pharmacological molecules or genetic tools targeting IRE1, PERK, ATF6 or any other UPR regulators, as well as their potential use in cancer models; and finally, (iii) novel preclinical results with UPR targeting drugs.

Dr. Eric Chevet
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

16 pages, 2378 KiB  
Article
Sigma 1 Receptor is Overexpressed in Hepatocellular Adenoma: Involvement of ERα and HNF1α
by Laure Villemain, Sylvie Prigent, Aurélie Abou-Lovergne, Laura Pelletier, Magali Chiral, Marco Pontoglio, Fabienne Foufelle, Stefano Caruso, Raphael Pineau, Sandra Rebouissou, Eric Chevet, Jessica Zucman-Rossi and Laurent Combettes
Cancers 2020, 12(8), 2213; https://doi.org/10.3390/cancers12082213 - 7 Aug 2020
Cited by 4 | Viewed by 2686
Abstract
Sigma receptor 1 (SigR1) is an endoplasmic reticulum resident integral membrane protein whose functions remain unclear. Although the liver shows the highest expression of SigR1, its role in this organ is unknown. SigR1 is overexpressed in many cancers and its expression is correlated [...] Read more.
Sigma receptor 1 (SigR1) is an endoplasmic reticulum resident integral membrane protein whose functions remain unclear. Although the liver shows the highest expression of SigR1, its role in this organ is unknown. SigR1 is overexpressed in many cancers and its expression is correlated to hormonal status in hormone-dependent cancers. To better understand the role of SigR1 in hepatocytes we focused our work on the regulation of its expression in tumoral liver. In this context, hepatocellular adenomas, benign hepatic tumors associated with estrogen intake are of particular interest. The expression of SigR1 mRNA was assessed in hepatocellular adenoma (HCA) patients using qPCR. The impact of estrogen on the expression of SigR1 was studied in vivo (mice) and in vitro (HepG2 and Huh7 cells). The effect of HNF1α on the expression of SigR1 was studied in vivo by comparing wild type mice to HNF1 knockout mice. Estrogen enhanced SigR1 expression through its nuclear receptor ERα. HNF1α mutated HCA (H-HCA) significantly overexpressed SigR1 compared to all other HCA subtypes. HNF1 knockout mice showed an increase in SigR1 expression. Overexpressing SigR1 in cellular models increases proliferation rate and storage of lipid droplets, which phenocopies the H-HCA phenotype. SigR1 is involved in hepatocyte proliferation and steatosis and may play an important role in the control of the H-HCA phenotype. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

13 pages, 1991 KiB  
Article
ATF4-Dependent NRF2 Transcriptional Regulation Promotes Antioxidant Protection during Endoplasmic Reticulum Stress
by Carmen Sarcinelli, Helena Dragic, Marie Piecyk, Virginie Barbet, Cédric Duret, Audrey Barthelaix, Carole Ferraro-Peyret, Joelle Fauvre, Toufic Renno, Cédric Chaveroux and Serge N. Manié
Cancers 2020, 12(3), 569; https://doi.org/10.3390/cancers12030569 - 1 Mar 2020
Cited by 63 | Viewed by 8282
Abstract
Endoplasmic reticulum (ER) stress generates reactive oxygen species (ROS) that induce apoptosis if left unabated. To limit oxidative insults, the ER stress PKR-like endoplasmic reticulum Kinase (PERK) has been reported to phosphorylate and activate nuclear factor erythroid 2-related factor 2 (NRF2). Here, we [...] Read more.
Endoplasmic reticulum (ER) stress generates reactive oxygen species (ROS) that induce apoptosis if left unabated. To limit oxidative insults, the ER stress PKR-like endoplasmic reticulum Kinase (PERK) has been reported to phosphorylate and activate nuclear factor erythroid 2-related factor 2 (NRF2). Here, we uncover an alternative mechanism for PERK-mediated NRF2 regulation in human cells that does not require direct phosphorylation. We show that the activation of the PERK pathway rapidly stimulates the expression of NRF2 through activating transcription factor 4 (ATF4). In addition, NRF2 activation is late and largely driven by reactive oxygen species (ROS) generated during late protein synthesis recovery, contributing to protecting against cell death. Thus, PERK-mediated NRF2 activation encompasses a PERK-ATF4-dependent control of NRF2 expression that contributes to the NRF2 protective response engaged during ER stress-induced ROS production. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

20 pages, 4396 KiB  
Article
The Expression of Myeloproliferative Neoplasm-Associated Calreticulin Variants Depends on the Functionality of ER-Associated Degradation
by Olivier Mansier, Valérie Prouzet-Mauléon, Gwénaële Jégou, Kim Barroso, Diana Pelizzari Raymundo, Aurélie Chauveau, Pierre-Yves Dumas, Valérie Lagarde, Béatrice Turcq, Jean-Max Pasquet, Jean-François Viallard, Chloé James, Vincent Praloran, Konstantinos Voutetakis, Aristotelis Chatziioannou, François-Xavier Mahon, Eric Chevet and Eric Lippert
Cancers 2019, 11(12), 1921; https://doi.org/10.3390/cancers11121921 - 2 Dec 2019
Cited by 7 | Viewed by 3850
Abstract
Background: Mutations in CALR observed in myeloproliferative neoplasms (MPN) were recently shown to be pathogenic via their interaction with MPL and the subsequent activation of the Janus Kinase – Signal Transducer and Activator of Transcription (JAK-STAT) pathway. However, little is known on the [...] Read more.
Background: Mutations in CALR observed in myeloproliferative neoplasms (MPN) were recently shown to be pathogenic via their interaction with MPL and the subsequent activation of the Janus Kinase – Signal Transducer and Activator of Transcription (JAK-STAT) pathway. However, little is known on the impact of those variant CALR proteins on endoplasmic reticulum (ER) homeostasis. Methods: The impact of the expression of Wild Type (WT) or mutant CALR on ER homeostasis was assessed by quantifying the expression level of Unfolded Protein Response (UPR) target genes, splicing of X-box Binding Protein 1 (XBP1), and the expression level of endogenous lectins. Pharmacological and molecular (siRNA) screens were used to identify mechanisms involved in CALR mutant proteins degradation. Coimmunoprecipitations were performed to define more precisely actors involved in CALR proteins disposal. Results: We showed that the expression of CALR mutants alters neither ER homeostasis nor the sensitivity of hematopoietic cells towards ER stress-induced apoptosis. In contrast, the expression of CALR variants is generally low because of a combination of secretion and protein degradation mechanisms mostly mediated through the ER-Associated Degradation (ERAD)-proteasome pathway. Moreover, we identified a specific ERAD network involved in the degradation of CALR variants. Conclusions: We propose that this ERAD network could be considered as a potential therapeutic target for selectively inhibiting CALR mutant-dependent proliferation associated with MPN, and therefore attenuate the associated pathogenic outcomes. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

Review

Jump to: Research

32 pages, 4078 KiB  
Review
Stress Management: Death Receptor Signalling and Cross-Talks with the Unfolded Protein Response in Cancer
by Elodie Lafont
Cancers 2020, 12(5), 1113; https://doi.org/10.3390/cancers12051113 - 29 Apr 2020
Cited by 11 | Viewed by 4582
Abstract
Throughout tumour progression, tumour cells are exposed to various intense cellular stress conditions owing to intrinsic and extrinsic cues, to which some cells are remarkably able to adapt. Death Receptor (DR) signalling and the Unfolded Protein Response (UPR) are two stress responses that [...] Read more.
Throughout tumour progression, tumour cells are exposed to various intense cellular stress conditions owing to intrinsic and extrinsic cues, to which some cells are remarkably able to adapt. Death Receptor (DR) signalling and the Unfolded Protein Response (UPR) are two stress responses that both regulate a plethora of outcomes, ranging from proliferation, differentiation, migration, cytokine production to the induction of cell death. Both signallings are major modulators of physiological tissue homeostasis and their dysregulation is involved in tumorigenesis and the metastastic process. The molecular determinants of the control between the different cellular outcomes induced by DR signalling and the UPR in tumour cells and their stroma and their consequences on tumorigenesis are starting to be unravelled. Herein, I summarize the main steps of DR signalling in relation to its cellular and pathophysiological roles in cancer. I then highlight how the UPR and DR signalling control common cellular outcomes and also cross-talk, providing potential opportunities to further understand the development of malignancies. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

17 pages, 1293 KiB  
Review
Alternative Mechanisms of p53 Action During the Unfolded Protein Response
by Leïla T. S. Fusée, Mónica Marín, Robin Fåhraeus and Ignacio López
Cancers 2020, 12(2), 401; https://doi.org/10.3390/cancers12020401 - 10 Feb 2020
Cited by 15 | Viewed by 4122
Abstract
The tumor suppressor protein p53 orchestrates cellular responses to a vast number of stresses, with DNA damage and oncogenic activation being some of the best described. The capacity of p53 to control cellular events such as cell cycle progression, DNA repair, and apoptosis, [...] Read more.
The tumor suppressor protein p53 orchestrates cellular responses to a vast number of stresses, with DNA damage and oncogenic activation being some of the best described. The capacity of p53 to control cellular events such as cell cycle progression, DNA repair, and apoptosis, to mention some, has been mostly linked to its role as a transcription factor. However, how p53 integrates different signaling cascades to promote a particular pathway remains an open question. One way to broaden its capacity to respond to different stimuli is by the expression of isoforms that can modulate the activities of the full-length protein. One of these isoforms is p47 (p53/47, Δ40p53, p53ΔN40), an alternative translation initiation variant whose expression is specifically induced by the PERK kinase during the Unfolded Protein Response (UPR) following Endoplasmic Reticulum stress. Despite the increasing knowledge on the p53 pathway, its activity when the translation machinery is globally suppressed during the UPR remains poorly understood. Here, we focus on the expression of p47 and we propose that the alternative initiation of p53 mRNA translation offers a unique condition-dependent mechanism to differentiate p53 activity to control cell homeostasis during the UPR. We also discuss how the manipulation of these processes may influence cancer cell physiology in light of therapeutic approaches. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

24 pages, 1444 KiB  
Review
The Unfolded Protein Response: A Novel Therapeutic Target in Acute Leukemias
by Alberto M. Martelli, Francesca Paganelli, Francesca Chiarini, Camilla Evangelisti and James A. McCubrey
Cancers 2020, 12(2), 333; https://doi.org/10.3390/cancers12020333 - 1 Feb 2020
Cited by 25 | Viewed by 6130
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved adaptive response triggered by the stress of the endoplasmic reticulum (ER) due, among other causes, to altered cell protein homeostasis (proteostasis). UPR is mediated by three main sensors, protein kinase RNA-like endoplasmic reticulum kinase [...] Read more.
The unfolded protein response (UPR) is an evolutionarily conserved adaptive response triggered by the stress of the endoplasmic reticulum (ER) due, among other causes, to altered cell protein homeostasis (proteostasis). UPR is mediated by three main sensors, protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor 6α (ATF6α), and inositol-requiring enzyme-1α (IRE1α). Given that proteostasis is frequently disregulated in cancer, UPR is emerging as a critical signaling network in controlling the survival, selection, and adaptation of a variety of neoplasias, including breast cancer, prostate cancer, colorectal cancer, and glioblastoma. Indeed, cancer cells can escape from the apoptotic pathways elicited by ER stress by switching UPR into a prosurvival mechanism instead of cell death. Although most of the studies on UPR focused on solid tumors, this intricate network plays a critical role in hematological malignancies, and especially in multiple myeloma (MM), where treatment with proteasome inhibitors induce the accumulation of unfolded proteins that severely perturb proteostasis, thereby leading to ER stress, and, eventually, to apoptosis. However, UPR is emerging as a key player also in acute leukemias, where recent evidence points to the likelihood that targeting UPR-driven prosurvival pathways could represent a novel therapeutic strategy. In this review, we focus on the oncogene-specific regulation of individual UPR signaling arms, and we provide an updated outline of the genetic, biochemical, and preclinical therapeutic findings that support UPR as a relevant, novel target in acute leukemias. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

30 pages, 857 KiB  
Review
Proteostasis in the Endoplasmic Reticulum: Road to Cure
by Su Min Nam and Young Joo Jeon
Cancers 2019, 11(11), 1793; https://doi.org/10.3390/cancers11111793 - 14 Nov 2019
Cited by 22 | Viewed by 4960
Abstract
The endoplasmic reticulum (ER) is an interconnected organelle that is responsible for the biosynthesis, folding, maturation, stabilization, and trafficking of transmembrane and secretory proteins. Therefore, cells evolve protein quality-control equipment of the ER to ensure protein homeostasis, also termed proteostasis. However, disruption in [...] Read more.
The endoplasmic reticulum (ER) is an interconnected organelle that is responsible for the biosynthesis, folding, maturation, stabilization, and trafficking of transmembrane and secretory proteins. Therefore, cells evolve protein quality-control equipment of the ER to ensure protein homeostasis, also termed proteostasis. However, disruption in the folding capacity of the ER caused by a large variety of pathophysiological insults leads to the accumulation of unfolded or misfolded proteins in this organelle, known as ER stress. Upon ER stress, unfolded protein response (UPR) of the ER is activated, integrates ER stress signals, and transduces the integrated signals to relive ER stress, thereby leading to the re-establishment of proteostasis. Intriguingly, severe and persistent ER stress and the subsequently sustained unfolded protein response (UPR) are closely associated with tumor development, angiogenesis, aggressiveness, immunosuppression, and therapeutic response of cancer. Additionally, the UPR interconnects various processes in and around the tumor microenvironment. Therefore, it has begun to be delineated that pharmacologically and genetically manipulating strategies directed to target the UPR of the ER might exhibit positive clinical outcome in cancer. In the present review, we summarize recent advances in our understanding of the UPR of the ER and the UPR of the ER–mitochondria interconnection. We also highlight new insights into how the UPR of the ER in response to pathophysiological perturbations is implicated in the pathogenesis of cancer. We provide the concept to target the UPR of the ER, eventually discussing the potential of therapeutic interventions for targeting the UPR of the ER for cancer treatment. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

28 pages, 2749 KiB  
Review
Leveraging the Role of the Metastatic Associated Protein Anterior Gradient Homologue 2 in Unfolded Protein Degradation: A Novel Therapeutic Biomarker for Cancer
by Reem Alsereihi, Hans-Juergen Schulten, Sherin Bakhashab, Kulvinder Saini, Ahmed M. Al-Hejin and Deema Hussein
Cancers 2019, 11(7), 890; https://doi.org/10.3390/cancers11070890 - 26 Jun 2019
Cited by 11 | Viewed by 4650
Abstract
Effective diagnostic, prognostic and therapeutic biomarkers can help in tracking disease progress, predict patients’ survival, and considerably affect the drive for successful clinical management. The present review aims to determine how the metastatic-linked protein anterior gradient homologue 2 (AGR2) operates to affect cancer [...] Read more.
Effective diagnostic, prognostic and therapeutic biomarkers can help in tracking disease progress, predict patients’ survival, and considerably affect the drive for successful clinical management. The present review aims to determine how the metastatic-linked protein anterior gradient homologue 2 (AGR2) operates to affect cancer progression, and to identify associated potential diagnostic, prognostic and therapeutic biomarkers, particularly in central nervous system (CNS) tumors. Studies that show a high expression level of AGR2, and associate the protein expression with the resilience to chemotherapeutic treatments or with poor cancer survival, are reported. The primary protein structures of the seven variants of AGR2, including their functional domains, are summarized. Based on experiments in various biological models, this review shows an orchestra of multiple molecules that regulate AGR2 expression, including a feedback loop with p53. The AGR2-associated molecular functions and pathways including genomic integrity, proliferation, apoptosis, angiogenesis, adhesion, migration, stemness, and inflammation, are detailed. In addition, the mechanisms that can enable the rampant oncogenic effects of AGR2 are clarified. The different strategies used to therapeutically target AGR2-positive cancer cells are evaluated in light of the current evidence. Moreover, novel associated pathways and clinically relevant deregulated genes in AGR2 high CNS tumors are identified using a meta-analysis approach. Full article
(This article belongs to the Special Issue Therapeutic Targeting of the Unfolded Protein Response in Cancer)
Show Figures

Figure 1

Back to TopTop