Network


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

Hotspot


Dive into the research topics where Maria I. Vaccaro is active.

Publication


Featured researches published by Maria I. Vaccaro.


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.


Autophagy | 2011

The emerging role of autophagy in the pathophysiology of diabetes mellitus

Claudio Gonzalez; Myung-Shik Lee; Piero Marchetti; Massimo Pietropaolo; Roberto Towns; Maria I. Vaccaro; Hirotaka Watada; John W. Wiley

An emerging body of evidence supports a role for autophagy in the pathophysiology of type 1 and type 2 diabetes mellitus. Persistent high concentrations of glucose lead to imbalances in the antioxidant capacity within the cell resulting in oxidative stress-mediated injury in both disorders. An anticipated consequence of impaired autophagy is the accumulation of dysfunctional organelles such as mitochondria within the cell. Mitochondria are the primary site of the production of reactive oxygen species (ROS), and an imbalance in ROS production relative to the cytoprotective action of autophagy may lead to the accumulation of ROS. Impaired mitochondrial function associated with increased ROS levels have been proposed as mechanisms contributing to insulin resistance. In this article we review and interpret the literature that implicates a role for autophagy in the pathophysiology of type 1 and type 2 diabetes mellitus as it applies to β-cell dysfunction, and more broadly to organ systems involved in complications of diabetes including the cardiovascular, renal and nervous systems.


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.


Molecular Biology of the Cell | 2008

The TP53INP2 Protein Is Required for Autophagy in Mammalian Cells

Jonathan Nowak; Cendrine Archange; J. Tardivel-Lacombe; Pierre Pontarotti; Marie-Josèphe Pébusque; Maria I. Vaccaro; Guillermo Velasco; Jean-Charles Dagorn; Juan L. Iovanna

Using a bioinformatic approach, we identified a TP53INP1-related gene encoding a protein with 30% identity with tumor protein 53-induced nuclear protein 1 (TP53INP1), which was named TP53INP2. TP53INP1 and TP53INP2 sequences were found in several species ranging from Homo sapiens to Drosophila melanogaster, but orthologues were found neither in earlier eukaryotes nor in prokaryotes. To gain insight into the function of the TP53INP2 protein, we carried out a yeast two-hybrid screening that showed that TP53INP2 binds to the LC3-related proteins GABARAP and GABARAP-like2, and then we demonstrated by coimmunoprecipitation that TP53INP2 interacts with these proteins, as well as with LC3 and with the autophagosome transmembrane protein VMP1. TP53INP2 translocates from the nucleus to the autophagosome structures after activation of autophagy by rapamycin or starvation. Also, we showed that TP53INP2 expression is necessary for autophagosome development because its small interfering RNA-mediated knockdown strongly decreases sensitivity of mammalian cells to autophagy. Finally, we found that interactions between TP53INP2 and LC3 or the LC3-related proteins GABARAP and GABARAP-like2 require autophagy and are modulated by wortmannin as judged by bioluminescence resonance energy transfer assays. We suggest that TP53INP2 is a scaffold protein that recruits LC3 and/or LC3-related proteins to the autophagosome membrane by interacting with the transmembrane protein VMP1. It is concluded that TP53INP2 is a novel gene involved in the autophagy of mammalian cells.


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.


The Journal of Pathology | 2003

The pancreatitis-associated protein induces lung inflammation in the rat through activation of TNFα expression in hepatocytes

Emma Folch-Puy; Andrés García-Movtero; Juan L. Iovanna; Jean Charles Dagorn; Neus Prats; Maria I. Vaccaro; Daniel Closa

The pancreatitis‐associated protein (PAP) is a pancreatic stress protein overexpressed during acute pancreatitis, a disease often accompanied by lung inflammation. We investigated whether PAP was involved in the occurrence of this remote complication of pancreatitis and whether the liver might be implicated in the process. PAP was injected into the vena cava of rats (40 or 400 µg/kg body weight). For comparison, pancreatitis was induced in rats by intraductal administration of sodium taurocholate. Three hours later, parameters of inflammation and mRNA concentrations of TNFα, P‐selectin, heat shock protein (HSP)‐70, and extracellular superoxide dismutase (EC‐SOD) were monitored in lung and liver. Significant increases in P‐selectin expression, neutrophil infiltration, and oxidative stress revealed that PAP treatment induced lung inflammation in rats and exacerbated inflammation in animals with pancreatitis. Plasma TNFα level was increased and TNFα mRNA was strongly overexpressed in liver, with concomitant activation of NF‐κB; in situ hybridization revealed that TNFα overexpression was mainly located to hepatocytes. Lung inflammation induced by PAP could be prevented by injection of anti‐TNFα antibodies. It was concluded that, during pancreatitis, PAP released by the pancreas could mediate lung inflammation through induction of hepatic TNFα expression and subsequent increase in circulating TNFα. Copyright


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.

Collaboration


Dive into the Maria I. Vaccaro's collaboration.

Top Co-Authors

Avatar

Alejandro Ropolo

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

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

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
Top Co-Authors

Avatar

Maria I. Molejon

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Osvaldo Tiscornia

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Romina Pardo

University of Buenos Aires

View shared research outputs
Researchain Logo
Decentralizing Knowledge