Network


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

Hotspot


Dive into the research topics where Alejandro Ropolo is active.

Publication


Featured researches published by Alejandro Ropolo.


Journal of Biological Chemistry | 2007

The Pancreatitis-induced Vacuole Membrane Protein 1 Triggers Autophagy in Mammalian Cells

Alejandro Ropolo; Daniel Grasso; Romina Pardo; Maria L. Sacchetti; Cendrine Archange; Andrea Lo Ré; Mylène Seux; Jonathan Nowak; Claudio Gonzalez; Juan L. Iovanna; Maria I. Vaccaro

Autophagy is a degradation process of cytoplasmic cellular constituents, which serves as a survival mechanism in starving cells, and it is characterized by sequestration of bulk cytoplasm and organelles in double-membrane vesicles called autophagosomes. Autophagy has been linked to a variety of pathological processes such as neurodegenerative diseases and tumorigenesis, which highlights its biological and medical importance. We have previously characterized the vacuole membrane protein 1 (VMP1) gene, which is highly activated in acute pancreatitis, a disease associated with morphological changes resembling autophagy. Here we show that VMP1 expression triggers autophagy in mammalian cells. VMP1 expression induces the formation of ultrastructural features of autophagy and recruitment of the microtubule-associated protein 1 light-chain 3 (LC3), which is inhibited after treatment with the autophagy inhibitor 3-methiladenine. VMP1 is induced by starvation and rapamycin treatments. Its expression is necessary for autophagy, because VMP1 small interfering RNA inhibits autophagosome formation under both autophagic stimuli. VMP1 is a transmembrane protein that co-localizes with LC3, a marker of the autophagosomes. It interacts with Beclin 1, a mammalian autophagy initiator, through the VMP1-Atg domain, which is essential for autophagosome formation. VMP1 endogenous expression co-localizes with LC3 in pancreas tissue undergoing pancreatitis-induced autophagy. Finally, VMP1 stable expression targeted to pancreas acinar cell in transgenic mice induces autophagosome formation. Our results identify VMP1 as a novel autophagy-related membrane protein involved in the initial steps of the mammalian cell autophagic process.


Journal of Biological Chemistry | 2011

Zymophagy, a Novel Selective Autophagy Pathway Mediated by VMP1-USP9x-p62, Prevents Pancreatic Cell Death

Daniel Grasso; Alejandro Ropolo; Andrea Lo Ré; Verónica Boggio; Maria I. Molejon; Juan L. Iovanna; Claudio Gonzalez; Raul Urrutia; Maria I. Vaccaro

Autophagy has recently elicited significant attention as a mechanism that either protects or promotes cell death, although different autophagy pathways, and the cellular context in which they occur, remain to be elucidated. We report a thorough cellular and biochemical characterization of a novel selective autophagy that works as a protective cell response. This new selective autophagy is activated in pancreatic acinar cells during pancreatitis-induced vesicular transport alteration to sequester and degrade potentially deleterious activated zymogen granules. We have coined the term “zymophagy” to refer to this process. The autophagy-related protein VMP1, the ubiquitin-protease USP9x, and the ubiquitin-binding protein p62 mediate zymophagy. Moreover, VMP1 interacts with USP9x, indicating that there is a close cooperation between the autophagy pathway and the ubiquitin recognition machinery required for selective autophagosome formation. Zymophagy is activated by experimental pancreatitis in genetically engineered mice and cultured pancreatic acinar cells and by acute pancreatitis in humans. Furthermore, zymophagy has pathophysiological relevance by controlling pancreatitis-induced intracellular zymogen activation and helping to prevent cell death. Together, these data reveal a novel selective form of autophagy mediated by the VMP1-USP9x-p62 pathway, as a cellular protective response.


Scientific Reports | 2013

The VMP1-Beclin 1 interaction regulates autophagy induction

Maria Inés Molejon; Alejandro Ropolo; Andrea Lo Ré; Verónica Boggio; Maria I. Vaccaro

The Vacuole Membrane Protein 1 -VMP1- is a pancreatitis-associated transmembrane protein whose expression triggers autophagy in several human diseases. In the current study, we unveil the mechanism through which this protein induces autophagosome formation in mammalian cells. We show that VMP1 autophagy-related function requires its 20-aminoacid C-terminus hydrophilic domain (VMP1-AtgD). This is achieved through its direct binding to the BH3 motif of Beclin 1 leading to the formation of a complex with the Class III phosphatidylinositol-3 kinase (PI3K) hVps34, a key positive regulator of autophagy, at the site where autophagosomes are generated. This interaction also concomitantly promotes the dissociation of Bcl-2, an autophagy inhibitor, from Beclin 1. Moreover, we show that the VMP1-Beclin 1-hVps34 complex favors the association of Atg16L1 and LC3 with the autophagosomal membranes. Collectively, these findings reveal that VMP1 expression recruits and activates the Class III PI3K complex at the site of autophagosome formation during mammalian autophagy.


Pancreatology | 2010

Gemcitabine Induces the VMP1-Mediated Autophagy Pathway to Promote Apoptotic Death in Human Pancreatic Cancer Cells

Romina Pardo; Andrea Lo Ré; Cendrine Archange; Alejandro Ropolo; Daniela L. Papademetrio; Claudio Gonzalez; Elida Alvarez; Juan L. Iovanna; Maria I. Vaccaro

Background/Aim: Autophagy is a degradation process of cytoplasmic cellular constituents. We have described the vacuole membrane protein-1 (VMP1) whose expression triggers autophagy in mammalian cells. The aim of this study was to analyze the role of autophagy in human pancreatic cancer cell death. Methods/Results: Here we show that gemcitabine, the standard chemotherapy for pancreatic cancer, induced autophagy in PANC-1 and MIAPaCa-2 cells, as evidenced by the accumulation of acidic vesicular organelles, the recruitment of microtubule-associated protein-1 light chain-3, and electron microscopy. In addition, gemcitabine treatment induced early expression of VMP1 in cancer cells. Gemcitabine also induced apoptosis detected by morphology, annexin V-positive cells, and cleavage of caspase-3. Surprisingly, 3-methyladenine, an autophagy inhibitor, decreased apoptosis in gemcitabine-treated cells, showing that autophagy leads to cancer cell apoptotic death. Finally, VMP1 knockdown decreased autophagy and apoptosis in gemcitabine-treated cancer cells. Conclusions: The VMP1-autophagy pathway promotes apoptosis in pancreatic cancer cells and mediates gemcitabine-induced cytotoxicity.


Journal of Biological Chemistry | 2012

Novel AKT1-GLI3-VMP1 pathway mediates KRAS oncogene-induced autophagy in cancer cells.

Andrea E. Lo Ré; Maite G. Fernandez-Barrena; Luciana L. Almada; Lisa D. Mills; Sherine F. Elsawa; George Lund; Alejandro Ropolo; Maria I. Molejon; Maria I. Vaccaro; Martin E. Fernandez-Zapico

Background: Autophagy plays a role in cancer development. Results: Oncogenic KRAS induces Vacuole Membrane Protein 1 (VMP1) through a novel AKT1-GLI3-p300 pathway and requires VMP1 to regulate autophagy in cancer cells. Conclusion: Define a novel pathway initiated by the oncogene KRAS regulating autophagy. Significance: These findings contribute to the understanding of the mechanism underlying oncogene-induced autophagy. Autophagy is an evolutionarily conserved degradation process of cytoplasmic cellular constituents. It has been suggested that autophagy plays a role in tumor promotion and progression downstream oncogenic pathways; however, the molecular mechanisms underlying this phenomenon have not been elucidated. Here, we provide both in vitro and in vivo evidence of a novel signaling pathway whereby the oncogene KRAS induces the expression of VMP1, a molecule needed for the formation of the authophagosome and capable of inducing autophagy, even under nutrient-replete conditions. RNAi experiments demonstrated that KRAS requires VMP1 to induce autophagy. Analysis of the mechanisms identified GLI3, a transcription factor regulated by the Hedgehog pathway, as an effector of KRAS signaling. GLI3 regulates autophagy as well as the expression and promoter activity of VMP1 in a Hedgehog-independent manner. Chromatin immunoprecipitation assays demonstrated that GLI3 binds to the VMP1 promoter and complexes with the histone acetyltransferase p300 to regulate promoter activity. Knockdown of p300 impaired KRAS- and GLI3-induced activation of this promoter. Finally, we identified the PI3K-AKT1 pathway as the signaling pathway mediating the expression and promoter activity of VMP1 upstream of the GLI3-p300 complex. Together, these data provide evidence of a new regulatory mechanism involved in autophagy that integrates this cellular process into the molecular network of events regulating oncogene-induced autophagy.


Autophagy | 2008

A novel mammalian trans-membrane protein reveals an alternative initiation pathway for autophagy

Maria I. Vaccaro; Alejandro Ropolo; Daniel Grasso; Juan L. Iovanna

Autophagy is an early cellular event during acute pancreatitis, a disease defined as pancreas self-digestion. The Vacuole Membrane Protein 1 (VMP1) is a trans-membrane protein highly activated in acinar cells early during pancreatitis-induced autophagy and it remains in the autophagosomal membrane. We have shown that VMP1 expression is able to trigger autophagy in mammalian cells, even under nutrient-replete conditions. VMP1 is induced by autophagy stimuli and its expression is required for autophagosome development. VMP1 interacts with Beclin 1 through its hydrophilic C-terminal region, which we named Atg domain, as it is essential for autophagy. Remarkably, VMP1 pancreas-specific transgenic expression in mice promotes autophagosome formation. Most of the autophagy-related proteins were described in yeast or have a yeast homologue. VMP1 does not have any known homologue in yeast but its expression is required to start the autophagic process in mammalian cells. These findings support the hypothesis that mammalian cells may regulate autophagy in a different way. We propose that VMP1 is a novel autophagy related trans-membrane protein, which may lead the way in the search for alternative mechanisms of autophagosome formation. Addendum to: Ropolo A, Grasso D, Pardo R, Sacchetti ML, Archange C, Re AL, Seux M, Nowak J, Gonzalez CD, Iovanna JL, Vaccaro MI. The pancreatitis-induced vacuole membrane protein 1 triggers autophagy in mammalian cells. J Biol Chem 2007; 282:37124-33.


Pancreatology | 2003

VMP1 expression correlates with acinar cell cytoplasmic vacuolization in arginine-induced acute pancreatitis

Maria I. Vaccaro; Daniel Grasso; Alejandro Ropolo; Juan L. Iovanna; M. Cristina Cerquetti

Background: Recently, we described the cloning of VMP1 (vacuole membrane protein 1). In vitro expression of VMP1 promotes formation of cytoplasmic vacuoles followed by cell death. In order to test if VMP1 expression is related to the cytoplasmic vacuolization of the acinar cells during acute pancreatitis, we studied the in vivo expression of the new gene during arginine-induced acute pancreatitis. Methods: Male Wistar rats injected with 500 mg/100 g of L-arginine were time-course sacrificed and pancreas tissue removed. Results: Northern blot analysis showed maximal induction of VMP1 after 24 h remaining high after 48 h of arginine administration. Significant increase in the number of TUNEL-stained cells were found at those periods. After 24 and 48 h of arginine administration, light micrographs from thin plastic toluidine blue sections revealed numerous vacuoles in the cytoplasm of acinar cells. In situ hybridization studies showed high expression of VMP1 in acinar cells with cytoplasmic vacuolization. VMP1 mRNA highly and significantly correlated with vacuole formation. Conclusion: These results suggest that VMP1 expression may be involved in the cytoplasmic vacuolization of acinar cells during the early stage of acute pancreatitis.


BioMed Research International | 2014

Autophagy, Warburg, and Warburg Reverse Effects in Human Cancer

Claudio Gonzalez; Silvia Alvarez; Alejandro Ropolo; Carla Rosenzvit; María Florencia González Bagnes; Maria I. Vaccaro

Autophagy is a highly regulated-cell pathway for degrading long-lived proteins as well as for clearing cytoplasmic organelles. Autophagy is a key contributor to cellular homeostasis and metabolism. Warburg hypothesized that cancer growth is frequently associated with a deviation of a set of energy generation mechanisms to a nonoxidative breakdown of glucose. This cellular phenomenon seems to rely on a respiratory impairment, linked to mitochondrial dysfunction. This mitochondrial dysfunction results in a switch to anaerobic glycolysis. It has been recently suggested that epithelial cancer cells may induce the Warburg effect in neighboring stromal fibroblasts in which autophagy was activated. These series of observations drove to the proposal of a putative reverse Warburg effect of pathophysiological relevance for, at least, some tumor phenotypes. In this review we introduce the autophagy process and its regulation and its selective pathways and role in cancer cell metabolism. We define and describe the Warburg effect and the newly suggested “reverse” hypothesis. We also discuss the potential value of modulating autophagy with several pharmacological agents able to modify the Warburg effect. The association of the Warburg effect in cancer and stromal cells to tumor-related autophagy may be of relevance for further development of experimental therapeutics as well as for cancer prevention.


Autophagy | 2013

VMP1 is a new player in the regulation of the autophagy-specific phosphatidylinositol 3-kinase complex activation

Maria Inés Molejon; Alejandro Ropolo; Maria I. Vaccaro

We have elucidated a novel mechanism through which the autophagy-specific class III phosphatidylinositol 3-kinase (PtdIns3K) complex can be recruited to the PAS in mammalian cells, through the interaction between BECN1 and the vacuole membrane protein 1 (VMP1), an integral autophagosomal membrane protein. This interaction involves the binding between the C-terminal 20 amino acids of the VMP1 hydrophilic domain, which we have named the VMP1 autophagy-related domain (VMP1-AtgD), and the BH3 domain of BECN1. The association between these two proteins allows the formation of the autophagy-specific PtdIns3K complex, which activity favors the generation of phosphatidylinositol-3-phosphate (PtdIns3P) and the subsequent association of the autophagy-related (ATG) proteins, including ATG16L1, with the phagophore membranes. Therefore, VMP1 regulates the PtdIns3K activity on the phagophore membrane through its interaction with BECN1. Our data provide a novel model describing one of the key steps in phagophore assembly site (PAS) formation and autophagy regulation, and positions VMP1 as a new interactor of the autophagy-specific PtdIns3K complex in mammalian cells.


Pancreatology | 2012

Chemotherapy and autophagy-mediated cell death in pancreatic cancer cells

Alejandro Ropolo; Claudia Bagnes; Maria I. Molejon; Andrea Lo Ré; Verónica Boggio; Claudio Gonzalez; Maria I. Vaccaro

Autophagy is an evolutionarily preserved degradation process of cytoplasmic cellular constituents and plays important physiological roles in human health and disease. It has been proposed that autophagy plays an important role both in tumor progression and in promotion of cancer cell death, although the molecular mechanisms responsible for this dual action of autophagy in cancer have not been elucidated. Pancreatic ductal adenocarcinoma is one of the most aggressive human malignancies with 2-3% five-year survival rate. Its poor prognosis has been attributed to the lack of specific symptoms and early detection tools, and its relatively refractory to traditional cytotoxic agents and radiotherapy. Experimental evidence pointed at autophagy as a pancreatic cancer cell mechanism to survive under adverse environmental conditions, or as a defective programmed cell death mechanism that favors pancreatic cancer cell resistance to treatment. Here, we consider several phenotypical alterations that have been related to increase or decrease the autophagic process in pancreatic tumor cells. We specially review autophagy as a cell death mechanism in response to chemotherapeutic drugs.

Collaboration


Dive into the Alejandro Ropolo's collaboration.

Top Co-Authors

Avatar

Maria I. Vaccaro

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Verónica Boggio

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Daniel Grasso

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Daniel Grasso

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Andrea Lo Ré

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Claudio Gonzalez

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Maria I. Molejon

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Romina Pardo

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Cintia Catrinacio

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge