Network


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

Hotspot


Dive into the research topics where Giuseppina Minopoli is active.

Publication


Featured researches published by Giuseppina Minopoli.


Journal of Biological Chemistry | 1998

The Fe65 Adaptor Protein Interacts through Its PID1 Domain with the Transcription Factor CP2/LSF/LBP1

Nicola Zambrano; Giuseppina Minopoli; Paola de Candia; Tommaso Russo

The neural protein Fe65 possesses three putative protein-protein interaction domains: one WW domain and two phosphotyrosine interaction/phosphotyrosine binding domains (PID1 and PID2); the most C-terminal of these domains (PID2) interacts in vivo with the Alzheimer’s β-amyloid precursor protein, whereas the WW domain binds to Mena, the mammalian homolog ofDrosophila-enabled protein. By the interaction trap procedure, we isolated a cDNA clone encoding a possible ligand of the N-terminal PID/PTB domain of Fe65 (PID1). Sequence analysis of this clone revealed that this ligand corresponded to the previously identified transcription factor CP2/LSF/LBP1. Co-immunoprecipitation experiments demonstrated that the interaction between Fe65 and CP2/LSF/LBP1 also takes place in vivo between the native molecules. The localization of both proteins was studied using fractionated cellular extracts. These experiments demonstrated that the various isoforms of CP2/LSF/LBP1 are differently distributed among subcellular fractions. At least one isoform, derived from alternative splicing (LSF-ID), is present outside the nucleus; Fe65 was found in both fractions. Furthermore, transfection experiments with an HA-tagged CP2/LSF/LBP1 cDNA demonstrated that Fe65 interacts also with the nuclear form of CP2/LSF/LBP1. Considering that the analysis of Fe65 distribution in fractionated cell extracts demonstrated that this protein is present both in nuclear and non-nuclear fractions, we examined the expression of Fe65 deletion mutants in the two fractions. This analysis allowed us to observe that a small region N-terminal to the WW domain is phosphorylated and is necessary for the presence of Fe65 in the nuclear fraction.


Journal of Biological Chemistry | 1997

Interaction of the Phosphotyrosine Interaction/Phosphotyrosine Binding-related Domains of Fe65 with Wild-type and Mutant Alzheimer's β-Amyloid Precursor Proteins

Nicola Zambrano; Joseph D. Buxbaum; Giuseppina Minopoli; Francesca Fiore; Paola de Candia; Stefano De Renzis; Raffaella Faraonio; Shasta L. Sabo; Jim Cheetham; Marius Sudol; Tommaso Russo

The two tandem phosphotyrosine interaction/phosphotyrosine binding (PID/PTB) domains of the Fe65 protein interact with the intracellular region of the Alzheimers β-amyloid precursor protein (APP). This interaction, previously demonstrated in vitro and in the yeast two hybrid system, also takes place in vivo in mammalian cells, as demonstrated here by anti-Fe65 co-immunoprecipitation experiments. This interaction differs from that occurring between other PID/PTB domain-containing proteins, such as Shc and insulin receptor substrate 1, and activated growth factor receptors as follows: (i) the Fe65-APP interaction is phosphorylation-independent; (ii) the region of the APP intracellular domain involved in the binding is larger than that of the growth factor receptor necessary for the formation of the complex with Shc; and (iii) despite a significant similarity the carboxyl-terminal regions of PID/PTB of Fe65 and of Shc are not functionally interchangeable in terms of binding cognate ligands. A role for Fe65 in the pathogenesis of familial Alzheimers disease is suggested by the finding that mutant APP, responsible for some cases of familial Alzheimers disease, shows an altered in vivo interaction with Fe65.


FEBS Letters | 1998

Fe65 and the protein network centered around the cytosolic domain of the Alzheimer's β-amyloid precursor protein

Tommaso Russo; Raffaella Faraonio; Giuseppina Minopoli; Paola de Candia; Stefano De Renzis; Nicola Zambrano

A distinctive tract of all the forms of Alzheimers disease is the extracellular deposition of a 40–42/43 amino acid‐long peptide derived from the so‐called β‐amyloid precursor protein (APP). This is a membrane protein of unknown function, whose short cytosolic domain has been recently demonstrated to interact with several proteins. One of these proteins, named Fe65, has the characteristics of an adaptor protein; in fact, it possesses three protein‐protein interaction domains: a WW domain and two PID/PTB domains. The interaction with APP requires the most C‐terminal PID/PTB domain, whereas the WW domain is responsible for the interaction with various proteins, one of which was demonstrated to be the mammalian homolog of the Drosophila enabled protein (Mena), which in turn interacts with the cytoskeleton. The second PID/PTB domain of Fe65 binds to the CP2/LSF/LBP1 protein, which is an already known transcription factor. The other proteins interacting with the cytosolic domain of APP are the Go heterotrimeric protein, APP‐BP1 and X11. The latter interacts with APP through a PID/PTB domain and possesses two other protein‐protein interaction domains. The small size of the APP cytodomain and the overlapping of its regions involved in the binding of Fe65 and X11 suggest the existence of competitive mechanisms regulating the binding of the various ligands to this cytosolic domain. In this short review the possible functional roles of this complex protein network and its involvement in the generation of Alzheimers phenotype are discussed.


Journal of Biological Chemistry | 2007

Essential Roles for Fe65, Alzheimer Amyloid Precursor-binding Protein, in the Cellular Response to DNA Damage

Giuseppina Minopoli; Maria Stante; Francesco Napolitano; Francesca Telese; Luigi Aloia; Mario De Felice; Roberto Di Lauro; Roberto Pacelli; Arturo Brunetti; Nicola Zambrano; Tommaso Russo

Fe65 interacts with the cytosolic domain of the Alzheimer amyloid precursor protein (APP). The functions of the Fe65 are still unknown. To address this point we generated Fe65 knockout (KO) mice. These mice do not show any obvious phenotype; however, when fibroblasts (mouse embryonic fibroblasts), isolated from Fe65 KO embryos, were exposed to low doses of DNA damaging agents, such as etoposide or H2O2, an increased sensitivity to genotoxic stress, compared with wild type animals, clearly emerged. Accordingly, brain extracts from Fe65 KO mice, exposed to non-lethal doses of ionizing radiations, showed high levels of γ-H2AX and p53, thus demonstrating a higher sensitivity to X-rays than wild type mice. Nuclear Fe65 is necessary to rescue the observed phenotype, and few minutes after the exposure of MEFs to DNA damaging agents, Fe65 undergoes phosphorylation in the nucleus. With a similar timing, the proteolytic processing of APP is rapidly affected by the genotoxic stress: in fact, the cleavage of the APP COOH-terminal fragments by γ-secretase is induced soon after the exposure of cells to etoposide, in a Fe65-dependent manner. These results demonstrate that Fe65 plays an essential role in the response of the cells to DNA damage.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Fe65 is required for Tip60-directed histone H4 acetylation at DNA strand breaks.

Maria Stante; Giuseppina Minopoli; Fabiana Passaro; Maddalena Raia; Luigi Del Vecchio; Tommaso Russo

Fe65 is a binding partner of the Alzheimers β-amyloid precursor protein APP. The possible involvement of this protein in the cellular response to DNA damage was suggested by the observation that Fe65 null mice are more sensitive to genotoxic stress than WT counterpart. Fe65 associated with chromatin under basal conditions and its involvement in DNA damage repair requires this association. A known partner of Fe65 is the histone acetyltransferase Tip60. Considering the crucial role of Tip60 in DNA repair, we explored the hypothesis that the phenotype of Fe65 null cells depended on its interaction with Tip60. We demonstrated that Fe65 knockdown impaired recruitment of Tip60-TRRAP complex to DNA double strand breaks and decreased histone H4 acetylation. Accordingly, the efficiency of DNA repair was decreased upon Fe65 suppression. To explore whether APP has a role in this mechanism, we analyzed a Fe65 mutant unable to bind to APP. This mutant failed to rescue the phenotypes of Fe65 null cells; furthermore, APP/APLP2 suppression results in the impairment of recruitment of Tip60-TRRAP complex to DNA double strand breaks, decreased histone H4 acetylation and repair efficiency. On these bases, we propose that Fe65 and its interaction with APP play an important role in the response to DNA damage by assisting the recruitment of Tip60-TRRAP to DNA damage sites.


Neurobiology of Disease | 2005

Interaction of Tau with Fe65 links tau to APP

Christian Barbato; Nadia Canu; Nicola Zambrano; Annalucia Serafino; Giuseppina Minopoli; Maria Teresa Ciotti; Giuseppina Amadoro; Tommaso Russo; Pietro Calissano

The beta-amyloid precursor protein APP and the microtubule-associated protein Tau play a crucial role in the pathogenesis of Alzheimers disease (AD). However, the possible molecular events linking these two proteins are still unknown. Here, we show that Fe65, one of the ligands of the APP cytodomain, is associated with Tau in vivo and in vitro, as demonstrated by co-immunoprecipitation, co-localization, and FRET experiments. Deletion studies indicated that the N-terminal domain of Tau and the PTB1 domain of Fe65 are required for this association. This interaction is regulated by the phosphorylation of Tau at selected sites, by glycogen synthase kinase-3beta (GSK3beta) and cyclin-dependent kinase 5 (Cdk5), and requires an intact microtubule network. Furthermore, laser scanner microscopy and co-immunoprecipitation experiments provide preliminary evidence of possible complex(es) involving Tau, Fe65, APP. These findings open new perspectives for the study of the possible crosstalk between these proteins in the pathogenesis of AD.


International Archives of Allergy and Immunology | 1999

Novel Autocrine and Paracrine Loops of the Stem Cell Factor/Chymase Network

Amato de Paulis; Giuseppina Minopoli; Fabrizio Dal Piaz; P. Pucci; Tommaso Russo; Gianni Marone

Background: The aim of this study was to investigate whether the secretory granules of human mast cells store stem cell factor (SCF). We also addressed the question whether mast cell chymase, a chymotrypsin–like protease, also present in the secretory granules of human mast cells cleaves SCF at the peptide bound between Phe 158 and Met159. Methods: The skin samples were obtained from patients with mastocytosis, undergoing skin biopsy for diagnostic purposes. Mast cells were isolated and purified from human lung parenchyma (human lung mast cells, HLMC) by countercurrent elutriation followed by discontinuous Percoll density gradient. SCF contents of human mast cells were assessed for immunoreactive SCF by ELISA. Western blot analysis of SCF and its cleavage products were performed with the MoAb anti–SCF 7H6. SCF and its proteolytic fragment were characterized by electrospray mass spectrometry (ES/MS). Results: SCF is present in the secretory granules of human skin and lung mast cells. Immunoreactive SCF (iSCF) was detected in the cell lysates of HLMC, but not in basophils. iSCF was rapidly (3 min) released after challenge with anti–IgE, and iSCF in supernatants rapidly declined after 30 min. ES/MS analysis of rhSCF1–166 treated with recombinant human chymase showed a polypeptide of 17,977.1±0.6 Da and a minor component of 697.4±0.1 Da generated by specific cleavage at Phe159. SCF1–166 and SCF1–159 similarly activated HLMC and potentiated anti–IgE–induced activation of these cells. The cleavage product SCF160–166 had no effect on mast cells. Western blot analysis of supernatants of anti–IgE activated HLMC incubated for various intervals with rhSCF1–166 showed that rhSCF1–166 was converted to a faster–migrating form with a molecular weight compatible with SCF1–159 and to several SCF species. Conclusion: SCF is stored in human mast cell secretory granules and is immunologically released by mast cells. SCF1–166 is rapidly cleaved by chymase and other proteases to several SCF species.


Cell Death & Differentiation | 2015

miR-23a, miR-24 and miR-27a protect differentiating ESCs from BMP4-induced apoptosis

Anna Musto; Angelica Navarra; A Vocca; Anna Gargiulo; Giuseppina Minopoli; Simona Romano; Maria Fiammetta Romano; Tommaso Russo; Silvia Parisi

Numerous studies have indicated that BMP4 signaling is involved in the regulation of the early steps of development. In mouse embryonic stem cells (ESCs), BMP4 is crucial to sustain pluripotency and blocks differentiation towards neural fate. Here, through a systematic analysis of miRNAs in ESCs, we establish that BMP4 signaling regulates miR-23a, 27a and 24-2, through the recruitment of phospho-Smads at the promoter of the gene encoding this miRNA cluster. Suppression of miR-23a/b, 27a/b and 24 does not affect self-renewal or pluripotency, but induces an evident change of ESC differentiation, with a significant increase of the cells undergoing apoptosis after the transition from ESCs to epiblast stem cells (EpiSCs). BMP4 has been previously reported to cause apoptosis during ESC differentiation. By blocking BMP4 signaling, we completely prevent the apoptosis induced by suppression of the miRs. This suggests that the effects of miR suppression are the result of enhanced BMP4 signaling. This hypothesis is further supported by the observation that Smad5, the transcription factor downstream of the BMP4 receptor, is targeted by the miRNAs of the 23a and 23b clusters. Altogether, our results highlight the existence of a regulatory loop, involving Smad5 and the miR-23a clusters, that modulates the apoptotic response of ESCs to BMP4.


Developmental Neurobiology | 2009

Notch activation induces neurite remodeling and functional modifications in SH-SY5Y neuronal cells.

Giulia Ferrari-Toninelli; Sara Anna Bonini; Daniela Uberti; Francesco Napolitano; Maria Stante; Federica Santoro; Giuseppina Minopoli; Nicola Zambrano; Tommaso Russo; Maurizio Memo

Notch proteins are definitely recognized as key regulators of the neuronal fate during embryo development, but their function in the adult brain is still largely unknown. We have previously demonstrated that Notch pathway stimulation increases microtubules stability followed by the remodeling of neuronal morphology with neurite varicosities loss, thicker neuritis, and enlarged growth cones. Here we show that the neurite remodeling is a dynamic event, dependent on transcription and translation, and with functional implications. Exposure of differentiated human SH‐SY5Y neuroblastoma cells to the Notch ligand Jagged1 induces varicosities loss all along the neurites, accompanied by the redistribution of presynaptic vesicles and the decrease in neurotransmitters release. As evaluated by time lapse digital imaging, dynamic changes in neurite morphology were rapidly reversible and dependent on the activation of the Notch signaling pathway. In fact, it was prevented by the inhibition of the proteolytic γ‐secretase enzyme or the transcription machinery, and was mimicked by the transfection of the intracellular domain of Notch. One hour after treatment with Jagged1, several genes were downregulated. Many of these genes encode proteins that are known to be involved in protein synthesis. These data suggest that in adult neurons, Notch pathway activates a transcriptional program that regulates the equilibrium between varicosities formation and varicosities loss in the neuronal presynaptic compartment involving the expression and redistribution of both structural and functional proteins.


International Journal of Molecular Sciences | 2013

miR-125b Regulates the Early Steps of ESC Differentiation through Dies1 in a TGF-Independent Manner

Marica Battista; Anna Musto; Angelica Navarra; Giuseppina Minopoli; Tommaso Russo; Silvia Parisi

Over the past few years, it has become evident that the distinctive pattern of miRNA expression seen in embryonic stem cells (ESCs) contributes to important signals in the choice of the cell fate. Thus, the identification of miRNAs and their targets, whose expression is linked to a specific step of differentiation, as well as the modulation of these miRNAs, may prove useful in the learning of how ESC potential is regulated. In this context, we have studied the expression profile of miRNAs during neural differentiation of ESCs. We have found that miR-125b is upregulated in the first steps of neural differentiation of ESCs. This miRNA targets the BMP4 co-receptor, Dies1, and, in turn, regulates the balance between BMP4 and Nodal/Activin signaling. The ectopic expression of miR-125b blocks ESC differentiation at the epiblast stage, and this arrest is rescued by restoring the expression of Dies1. Finally, opposite to miR-125a, whose expression is under the control of the BMP4, miR-125b is not directly regulated by Transforming Growth Factor beta (TGFβ) signals. These results highlight a new important role of miR-125b in the regulation of the transition from ESCs to the epiblast stage and add a new level of control on TGFβ signaling in ESCs.

Collaboration


Dive into the Giuseppina Minopoli's collaboration.

Top Co-Authors

Avatar

Tommaso Russo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Nicola Zambrano

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Gargiulo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Chiara Stella Di Stadio

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Emilia Rippa

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Filomena Altieri

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Giuseppina Miselli

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paolo Arcari

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge