Network


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

Hotspot


Dive into the research topics where Hery Urra is active.

Publication


Featured researches published by Hery Urra.


Biochimica et Biophysica Acta | 2013

When ER stress reaches a dead end

Hery Urra; Estefanie Dufey; Fernanda Lisbona; Diego Rojas-Rivera; Claudio Hetz

Endoplasmic reticulum (ER) stress is a common feature of several physiological and pathological conditions affecting the function of the secretory pathway. To restore ER homeostasis, an orchestrated signaling pathway is engaged that is known as the unfolded protein response (UPR). The UPR has a primary function in stress adaptation and cell survival; however, under irreversible ER stress a switch to pro-apoptotic signaling events induces apoptosis of damaged cells. The mechanisms that initiate ER stress-dependent apoptosis are not fully understood. Several pathways have been described where we highlight the participation of the BCL-2 family of proteins and ER calcium release. In addition, recent findings also suggest that microRNAs and oxidative stress are relevant players on the transition from adaptive to cell death programs. Here we provide a global and integrated overview of the signaling networks that may determine the elimination of a cell under chronic ER stress. This article is part of a Special Section entitled: Cell Death Pathways.


The EMBO Journal | 2012

BH3‐only proteins are part of a regulatory network that control the sustained signalling of the unfolded protein response sensor IRE1α

Diego A. Rodriguez; Sebastian Zamorano; Fernanda Lisbona; Diego Rojas-Rivera; Hery Urra; Juan R Cubillos-Ruiz; Ricardo Armisen; Daniel R. Henriquez; Emily H. Cheng; Michal Letek; Tomas Vaisar; Thergiory Irrazabal; Christian González-Billault; Anthony Letai; Felipe X. Pimentel-Muiños; Guido Kroemer; Claudio Hetz

Adaptation to endoplasmic reticulum (ER) stress depends on the activation of the unfolded protein response (UPR) stress sensor inositol‐requiring enzyme 1α (IRE1α), which functions as an endoribonuclease that splices the mRNA of the transcription factor XBP‐1 (X‐box‐binding protein‐1). Through a global proteomic approach we identified the BCL‐2 family member PUMA as a novel IRE1α interactor. Immun oprecipitation experiments confirmed this interaction and further detected the association of IRE1α with BIM, another BH3‐only protein. BIM and PUMA double‐knockout cells failed to maintain sustained XBP‐1 mRNA splicing after prolonged ER stress, resulting in early inactivation. Mutation in the BH3 domain of BIM abrogated the physical interaction with IRE1α, inhibiting its effects on XBP‐1 mRNA splicing. Unexpectedly, this regulation required BCL‐2 and was antagonized by BAD or the BH3 domain mimetic ABT‐737. The modulation of IRE1α RNAse activity by BH3‐only proteins was recapitulated in a cell‐free system suggesting a direct regulation. Moreover, BH3‐only proteins controlled XBP‐1 mRNA splicing in vivo and affected the ER stress‐regulated secretion of antibodies by primary B cells. We conclude that a subset of BCL‐2 family members participates in a new UPR‐regulatory network, thus assuming apoptosis‐unrelated functions.


Trends in cancer | 2016

Endoplasmic Reticulum Stress and the Hallmarks of Cancer

Hery Urra; Estefanie Dufey; Tony Avril; Eric Chevet; Claudio Hetz

Tumor cells are often exposed to intrinsic and external factors that alter protein homeostasis, thus producing endoplasmic reticulum (ER) stress. To cope with this, cells evoke an adaptive mechanism to restore ER proteostasis known as the unfolded protein response (UPR). The three main UPR signaling branches initiated by IRE1α, PERK, and ATF6 are crucial for tumor growth and aggressiveness as well as for microenvironment remodeling or resistance to treatment. We provide a comprehensive overview of the contribution of the UPR to cancer biology and the acquisition of malignant characteristics, thus highlighting novel aspects including inflammation, invasion and metastasis, genome instability, resistance to chemo/radiotherapy, and angiogenesis. The therapeutic potential of targeting ER stress signaling in cancer is also discussed.


Science Signaling | 2014

Interplay Between the Oxidoreductase PDIA6 and microRNA-322 Controls the Response to Disrupted Endoplasmic Reticulum Calcium Homeostasis

Jody Groenendyk; Zhenling Peng; Elzbieta Dudek; Xiao Fan; Marcin J. Mizianty; Estefanie Dufey; Hery Urra; Denisse Sepulveda; Diego Rojas-Rivera; Yunki Lim; Do Han Kim; Kayla Baretta; Sonal Srikanth; Yousang Gwack; Joohong Ahnn; Randal J. Kaufman; Sun-Kyung Lee; Claudio Hetz; Lukasz Kurgan; Marek Michalak

Depletion of Ca2+ in the endoplasmic reticulum favors activation of a stress response involving IRE1α. Responding the Right Way to Cellular Stress Some proteins must be folded correctly in the endoplasmic reticulum (ER) to function properly. Various stress conditions can cause the buildup of unfolded proteins in the ER, which can cause cell death. There are multiple ways in which cells can respond to deal with the buildup of unfolded proteins. Groenendyk et al. investigated how cells deal with the stress of depletion of calcium ions from the ER and identified a pathway involving a microRNA and an oxidoreductase in the ER. They found that depletion of calcium from the ER resulted in the decreased abundance of a microRNA, which enabled a target mRNA and the oxidoreductase it encoded to accumulate. The oxidoreductase then activated a specific stress response. The authors showed that this pathway could be present in mice and nematodes. The disruption of the energy or nutrient balance triggers endoplasmic reticulum (ER) stress, a process that mobilizes various strategies, collectively called the unfolded protein response (UPR), which reestablish homeostasis of the ER and cell. Activation of the UPR stress sensor IRE1α (inositol-requiring enzyme 1α) stimulates its endoribonuclease activity, leading to the generation of the mRNA encoding the transcription factor XBP1 (X-box binding protein 1), which regulates the transcription of genes encoding factors involved in controlling the quality and folding of proteins. We found that the activity of IRE1α was regulated by the ER oxidoreductase PDIA6 (protein disulfide isomerase A6) and the microRNA miR-322 in response to disruption of ER Ca2+ homeostasis. PDIA6 interacted with IRE1α and enhanced IRE1α activity as monitored by phosphorylation of IRE1α and XBP1 mRNA splicing, but PDIA6 did not substantially affect the activity of other pathways that mediate responses to ER stress. ER Ca2+ depletion and activation of store-operated Ca2+ entry reduced the abundance of the microRNA miR-322, which increased PDIA6 mRNA stability and, consequently, IRE1α activity during the ER stress response. In vivo experiments with mice and worms showed that the induction of ER stress correlated with decreased miR-322 abundance, increased PDIA6 mRNA abundance, or both. Together, these findings demonstrated that ER Ca2+, PDIA6, IRE1α, and miR-322 function in a dynamic feedback loop modulating the UPR under conditions of disrupted ER Ca2+ homeostasis.


PLOS ONE | 2012

Caveolin-1-Enhanced Motility and Focal Adhesion Turnover Require Tyrosine-14 but Not Accumulation to the Rear in Metastatic Cancer Cells

Hery Urra; Vicente A. Torres; Rina Ortiz; Lorena Lobos; María Inés Díaz; Natalia Díaz; Steffen Härtel; Lisette Leyton; Andrew F.G. Quest

Caveolin-1 is known to promote cell migration, and increased caveolin-1 expression is associated with tumor progression and metastasis. In fibroblasts, caveolin-1 polarization and phosphorylation of tyrosine-14 are essential to promote migration. However, the role of caveolin-1 in migration of metastatic cells remains poorly defined. Here, caveolin-1 participation in metastatic cell migration was evaluated by shRNA targeting of endogenous caveolin-1 in MDA-MB-231 human breast cancer cells and ectopic expression in B16-F10 mouse melanoma cells. Depletion of caveolin-1 in MDA-MB-231 cells reduced, while expression in B16-F10 cells promoted migration, polarization and focal adhesion turnover in a sequence of events that involved phosphorylation of tyrosine-14 and Rac-1 activation. In B16-F10 cells, expression of a non-phosphorylatable tyrosine-14 to phenylalanine mutant failed to recapitulate the effects observed with wild-type caveolin-1. Alternatively, treatment of MDA-MB-231 cells with the Src family kinase inhibitor PP2 reduced caveolin-1 phosphorylation on tyrosine-14 and cell migration. Surprisingly, unlike for fibroblasts, caveolin-1 polarization and re-localization to the trailing edge were not observed in migrating metastatic cells. Thus, expression and phosphorylation, but not polarization of caveolin-1 favor the highly mobile phenotype of metastatic cells.


Pigment Cell & Melanoma Research | 2013

E-cadherin determines Caveolin-1 tumor suppression or metastasis enhancing function in melanoma cells

Lorena Lobos-González; Lorena Aguilar; Jorge Díaz; Natalia Díaz; Hery Urra; Vicente A. Torres; Veronica Silva; Christopher Fitzpatrick; Alvaro Lladser; Keith S. Hoek; Lisette Leyton; Andrew F.G. Quest

The role of caveolin‐1 (CAV1) in cancer is highly controversial. CAV1 suppresses genes that favor tumor development, yet also promotes focal adhesion turnover and migration of metastatic cells. How these contrasting observations relate to CAV1 function in vivo is unclear. Our previous studies implicate E‐cadherin in CAV1‐dependent tumor suppression. Here, we use murine melanoma B16F10 cells, with low levels of endogenous CAV1 and E‐cadherin, to unravel how CAV1 affects tumor growth and metastasis and to assess how co‐expression of E‐cadherin modulates CAV1 function in vivo in C57BL/6 mice. We find that overexpression of CAV1 in B16F10 (cav‐1) cells reduces subcutaneous tumor formation, but enhances metastasis relative to control cells. Furthermore, E‐cadherin expression in B16F10 (E‐cad) cells reduces subcutaneous tumor formation and lung metastasis when intravenously injected. Importantly, co‐expression of CAV1 and E‐cadherin in B16F10 (cav‐1/E‐cad) cells abolishes tumor formation, lung metastasis, increased Rac‐1 activity, and cell migration observed with B16F10 (cav‐1) cells. Finally, consistent with the notion that CAV1 participates in switching human melanomas to a more malignant phenotype, elevated levels of CAV1 expression correlated with enhanced migration and Rac‐1 activation in these cells.


Science Signaling | 2017

Endoplasmic reticulum proteostasis in glioblastoma—From molecular mechanisms to therapeutic perspectives

Joanna Obacz; Tony Avril; Pierre-Jean Le Reste; Hery Urra; Véronique Quillien; Claudio Hetz; Eric Chevet

Combined therapies targeting the unfolded protein stress response might be a way to treat glioblastomas. Gloss Glioblastoma (GBM) is the most common and aggressive brain tumor. Standard care combines surgery, radiotherapy, and chemotherapy, but patient median survival does not exceed 15 months. The unfolded protein response (UPR) is an adaptive cellular signaling pathway that promotes restoration of endoplasmic reticulum proteostasis. The UPR plays instrumental roles in various cancers, particularly growth, invasion, therapeutic resistance, and angiogenesis in GBM. In this Review, which contains four figures, two tables, and 219 references, we discuss how adjuvant or neoadjuvant UPR-targeted compounds could impede GBM growth and increase the efficacy of current treatments. Cellular stress induced by the accumulation of misfolded proteins at the endoplasmic reticulum (ER) is a central feature of secretory cells and is observed in many tissues in various diseases, including cancer, diabetes, obesity, and neurodegenerative disorders. Cellular adaptation to ER stress is achieved by the activation of the unfolded protein response (UPR), an integrated signal transduction pathway that transmits information about the protein folding status at the ER to the cytosol and nucleus to restore proteostasis. In the past decade, ER stress has emerged as a major pathway in remodeling gene expression programs that either prevent transformation or provide selective advantage in cancer cells. Controlled by the formation of a dynamic scaffold onto which many regulatory components assemble, UPR signaling is a highly regulated process that leads to an integrated reprogramming of the cell. In this Review, we provide an overview of the regulatory mechanisms underlying UPR signaling and how this pathway modulates cancer progression, particularly the aggressiveness and chemotherapeutic resistance exhibited by glioblastoma, a form of brain cancer. We also discuss the emerging cross-talk between the UPR and related metabolic processes to ensure maintenance of proteostasis, and we highlight possible therapeutic opportunities for targeting the pathway with small molecules.


The Journal of Infectious Diseases | 2010

Helicobacter pylori—Induced Loss of the Inhibitor-of-Apoptosis Protein Survivin Is Linked to Gastritis and Death of Human Gastric Cells

Manuel Valenzuela; Guillermo I. Perez-Perez; Alejandro H. Corvalán; Gonzalo Carrasco; Hery Urra; Denisse Bravo; Héctor Toledo; Andrew F.G. Quest

Helicobacter pylori infects the human stomach and modifies signaling pathways that affect gastric epithelial cell proliferation and viability. Chronic exposure to this pathogen contributes to the onset of gastric atrophy, an early event in the genesis of gastric cancer associated with H. pylori infection. Susceptibility to H. pylori-induced cell death ultimately depends on the presence of protective host cell factors. Although expression of the inhibitor-of-apoptosis protein survivin in adults is frequently linked to the development of cancer, evidence indicating that the protein is present in normal gastric mucosa is also available. Thus, we investigated in human gastric tissue samples and cell lines whether H. pylori infection is linked to loss of survivin and increased cell death. Our results show that infection with H. pylori decreased survivin protein levels in the mucosa of patients with gastritis. Furthermore, survivin down-regulation correlated with apoptosis and loss of cell viability in gastrointestinal cells cocultured with different H. pylori strains. Finally, overexpression of survivin in human gastric cells was sufficient to reduce cell death after infection. Taken together, these findings implicate survivin as an important survival factor in the gastric mucosa of humans.


Seminars in Cancer Biology | 2015

ER proteostasis addiction in cancer biology: Novel concepts.

Estefanie Dufey; Hery Urra; Claudio Hetz

Endoplasmic reticulum (ER) stress is generated by various physiological and pathological conditions that induce an accumulation of misfolded proteins in its lumen. ER stress activates the unfolded protein response (UPR), an adaptive reaction to cope with protein misfolding to and restore proteostasis. However, chronic ER stress results in apoptosis. In solid tumors, the UPR mediates adaptation to various environmental stressors, including hypoxia, low in pH and low nutrients availability, driving positive selection. Recent findings support the concept that UPR signaling also contributes to other relevant cancer-related event that may not be related to ER stress, including angiogenesis, genomic instability, metastasis and immunomodulation. In this article, we overview novel discoveries highlighting the impact of the UPR to different aspects of cancer biology beyond its known role as a survival factor to the hypoxic environment observed in solid tumors.


Molecular Cell | 2018

Interactome Screening Identifies the ER Luminal Chaperone Hsp47 as a Regulator of the Unfolded Protein Response Transducer IRE1α

Denisse Sepulveda; Diego Rojas-Rivera; Diego A. Rodriguez; Jody Groenendyk; Andrés Kohler; Cynthia Lebeaupin; Shinya Ito; Hery Urra; Amado Carreras-Sureda; Younis M. Hazari; Mireille Vasseur-Cognet; Maruf M.U. Ali; Eric Chevet; Gisela Campos; Patricio Godoy; Tomas Vaisar; Béatrice Bailly-Maitre; Kazuhiro Nagata; Marek Michalak; Jimena Sierralta; Claudio Hetz

Maintenance of endoplasmic reticulum (ER) proteostasis is controlled by a dynamic signaling network known as the unfolded protein response (UPR). IRE1α is a major UPR transducer, determining cell fate under ER stress. We used an interactome screening to unveil several regulators of the UPR, highlighting the ER chaperone Hsp47 as the major hit. Cellular and biochemical analysis indicated that Hsp47 instigates IRE1α signaling through a physical interaction. Hsp47 directly binds to the ER luminal domain of IRE1α with high affinity, displacing the negative regulator BiP from the complex to facilitate IRE1α oligomerization. The regulation of IRE1α signaling by Hsp47 is evolutionarily conserved as validated using fly and mouse models of ER stress. Hsp47 deficiency sensitized cells and animals to experimental ER stress, revealing the significance of Hsp47 to global proteostasis maintenance. We conclude that Hsp47 adjusts IRE1α signaling by fine-tuning the threshold to engage an adaptive UPR.

Collaboration


Dive into the Hery Urra's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge