Network


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

Hotspot


Dive into the research topics where Carlotta Zamparelli is active.

Publication


Featured researches published by Carlotta Zamparelli.


FEBS Letters | 1995

Calcium-dependent translocation of sorcin to membranes: functional relevance in contractile tissue.

Marian B. Meyers; Carlotta Zamparelli; Daniela Verzili; Adam P. Dicker; Thomas J.J. Blanck; Emilia Chiancone

Sorcin, a 22 kDa calcium binding protein present in abundance in cardiac tissue and in multi‐drug resistant cells and previously described as a soluble protein, is now shown to undergo a calcium‐dependent translocation process from the cytosol to cellular membranes in both systems. The translocation process takes place also in E. coli BL21 cells that express recombinant sorcin, r‐sorcin, and can be exploited in the purification of the protein. Calcium binding to purified r‐sorcin occurs at micromolar concentrations of the metal and is accompanied by a conformational change that renders the protein soluble in the non‐ionic detergent Triton X‐114. This finding suggests that lipids are the target of sorcin on cellular membranes. The possible significance of the calcium‐dependent translocation of sorcin in the specialized functions of sorcin‐expressing cells is discussed.


Nanoscale | 2013

Antibody–drug conjugates: targeting melanoma with cisplatin encapsulated in protein-cage nanoparticles based on human ferritin

Elisabetta Falvo; Elisa Tremante; Rocco Fraioli; Carlo Leonetti; Carlotta Zamparelli; Alberto Boffi; Veronica Morea; Pierpaolo Ceci; Patrizio Giacomini

A novel antibody-drug conjugate (ADC) was synthesized incorporating ferritin-based nanoparticles. An average of three molecules of monoclonal antibody (mAb) Ep1 to the human melanoma-specific antigen CSPG4 were conjugated to a single ferritin cage encapsulating about 50 cisplatin molecules (HFt-Pt-Ep1). The HFt-Pt-Ep1 nanoparticle had an estimated molecular size of about 900 kD and 33 nm, and flow cytometry demonstrated specific binding to a CSPG4(+) melanoma cell line, but not to a CSPG4(-) breast carcinoma cell line. As compared to the cisplatin-containing ferritin nanoparticle alone (HFt-Pt), which inhibited thymidine incorporation more efficiently in breast carcinoma than melanoma cells, the mAb-derivatized HFt-Pt-Ep1 nanoparticle had a 25-fold preference for the latter. A similar preference for melanoma was observed upon systemic intravenous administration of HFt-Pt-Ep1 to nude mice xenotransplanted with pre-established, palpable melanoma and breast carcinoma tumors. Thus, we have been able to determine precise combinations and stoichiometric relationships between mAbs and nanoparticle protein cages, whereby the latter lose their tropism for ubiquitously distributed cellular receptors, and acquire instead remarkably lineage-selective binding. HFt-Pt-Ep1 is therefore an interesting model to improve the therapeutic index of antiblastic therapy in a tumor such as melanoma, which at its advanced stages is totally refractory to mono- and combination-chemotherapy.


Journal of Muscle Research and Cell Motility | 1999

Expression and localisation of annexin VII (synexin) isoforms in differentiating myoblasts.

Christoph S. Clemen; Andreas Hofmann; Carlotta Zamparelli; Angelika A. Noegel

Annexin VII exists in a 47 kDa and a 51 kDa isoform with the 51 kDa protein being the only isoform present in skeletal muscle. Expression of the 51 kDa isoform during myogenesis and localization was studied in cells after conversion into myogenic cells by transduction with MyoD and in mouse and human myogenic cell lines. MyoD expression in NIH3T3 and C3H10T1/2 fibroblasts led to disappearance of the mRNA specific for the 47kDa isoform and appearance of the 51 kDa isoform-specific mRNA. The overall amount of annexin VII protein was reduced in myogenic converted cells. Both in undifferentiated and differentiated cells annexin VII was localized by immunofluorescence microscopy to punctate structures which were distributed all over the cell. A GFP annexin VII fusion protein showed a similar distribution. Cell fractionation studies indicated that annexin VII is equally distributed between cytosol and membrane fractions in undifferentiated cells, while in differentiated cells it is exclusively present in the membrane fraction. By sucrose gradient centrifugation of postnuclear supernatants we identified two distinct annexin VII-containing membrane populations that cofractionated with caveolin 3- and sorcin-containing membranes.


Journal of Biological Chemistry | 2009

Iron translocation into and out of Listeria innocua Dps and size distribution of the protein-enclosed nanomineral are modulated by the electrostatic gradient at the 3-fold "ferritin-like" pores.

Giuliano Bellapadrona; Simonetta Stefanini; Carlotta Zamparelli; Elizabeth C. Theil; Emilia Chiancone

Elucidating pore function at the 3-fold channels of 12-subunit, microbial Dps proteins is important in understanding their role in the management of iron/hydrogen peroxide. The Dps pores are called “ferritin-like” because of the structural resemblance to the 3-fold channels of 24-subunit ferritins used for iron entry and exit to and from the protein cage. In ferritins, negatively charged residues lining the pores generate a negative electrostatic gradient that guides iron ions toward the ferroxidase centers for catalysis with oxidant and destined for the mineralization cavity. To establish whether the set of three aspartate residues that line the pores in Listeria innocua Dps act in a similar fashion, D121N, D126N, D130N, and D121N/D126N/D130N proteins were produced; kinetics of iron uptake/release and the size distribution of the iron mineral in the protein cavity were compared. The results, discussed in the framework of crystal growth in a confined space, indicate that iron uses the hydrophilic 3-fold pores to traverse the protein shell. For the first time, the strength of the electrostatic potential is observed to modulate kinetic cooperativity in the iron uptake/release processes and accordingly the size distribution of the microcrystalline iron minerals in the Dps protein population.


FEBS Letters | 2000

The sorcin-annexin VII calcium-dependent interaction requires the sorcin N-terminal domain

Daniela Verzili; Carlotta Zamparelli; Benedetta Mattei; Angelika A. Noegel; Emilia Chiancone

Surface plasmon resonance experiments show that at neutral pH the stability of the complex between sorcin and annexin VII (synexin) increases dramatically between 3 and 6 μM calcium; at the latter cation concentration the K D value is 0.63 μM. In turn, the lack of complex formation between the sorcin Ca2+ binding domain (33–198) and synexin maps the annexin binding site to the N‐terminal region of the sorcin polypeptide chain. Annexin VII likewise employs the N‐terminal domain, more specifically the first 31 amino acids, to interact with sorcin [Brownawell, A.M. and Creutz, C.E. (1997) J. Biol. Chem. 272, 22182–22190]. The interaction may involve similar structural motifs in the two proteins, namely GGYY and GYGG in sorcin and GYPP in synexin.


BMC Biochemistry | 2003

Function, expression and localization of annexin A7 in platelets and red blood cells: Insights derived from an annexin A7 mutant mouse

Claudia Herr; Christoph S. Clemen; Gisela Lehnert; Rüdiger Kutschkow; Susanne M. Picker; Birgit S. Gathof; Carlotta Zamparelli; Michael Schleicher; Angelika A. Noegel

BackgroundAnnexin A7 is a Ca2+- and phospholipid-binding protein expressed as a 47 and 51 kDa isoform, which is thought to be involved in membrane fusion processes. Recently the 47 kDa isoform has been identified in erythrocytes where it was proposed to be a key component in the process of the Ca2+-dependent vesicle release, a process with which red blood cells might protect themselves against an attack by for example complement components.ResultsThe role of annexin A7 in red blood cells was addressed in erythrocytes from anxA7-/- mice. Interestingly, the Ca2+-mediated vesiculation process was not impaired. Also, the membrane organization appeared not to be disturbed as assessed using gradient fractionation studies. Instead, lack of annexin A7 led to an altered cell shape and increased osmotic resistance of red blood cells. Annexin A7 was also identified in platelets. In these cells its loss led to a slightly slower aggregation velocity which seems to be compensated by an increased number of platelets. The results appear to rule out an important role of annexin A7 in membrane fusion processes occurring in red blood cells. Instead the protein might be involved in the organization of the membrane cytoskeleton. Red blood cells may represent an appropriate model to study the role of annexin A7 in cellular processes.ConclusionWe have demonstrated the presence of both annexin A7 isoforms in red blood cells and the presence of the small isoform in platelets. In both cell types the loss of annexin A7 impairs cellular functions. The defects observed are however not compatible with a crucial role for annexin A7 in membrane fusion processes in these cell types.


FEBS Letters | 1997

CALCIUM- AND PH-LINKED OLIGOMERIZATION OF SORCIN CAUSING TRANSLOCATION FROM CYTOSOL TO MEMBRANES

Carlotta Zamparelli; Andrea Ilari; Daniela Verzili; Paola Vecchini; Emilia Chiancone

Sorcin, a cytosolic calcium‐binding protein containing a pair of EF‐hand motifs, undergoes a Ca2+‐dependent translocation to the cell membrane. The underlying conformational change is similar at pH 6.0 and 7.5 and consists in an increase in overall hydrophobicity that involves the aromatic residues and in particular the two tryptophan residues which become less exposed to solvent. The concomitant association from dimers to tetramers indicates that the tryptophan residues, which are located between the EF‐hand sites, become buried at the dimer–dimer interface. Ca2+‐bound sorcin displays a striking difference in solubility as a function of pH that has been ascribed to the formation of calcium‐stabilized aggregates.


Journal of Molecular and Cellular Cardiology | 2010

Activation of the cardiac Na+–Ca2+ exchanger by sorcin via the interaction of the respective Ca2+-binding domains

Carlotta Zamparelli; Niall Macquaide; Gianni Colotti; Daniela Verzili; Tim Seidler; Godfrey L. Smith; Emilia Chiancone

Sorcin is a penta-EF-hand protein that interacts with intracellular target proteins after Ca2+ binding. The sarcolemmal Na+/Ca2+ exchanger (NCX1) may be an important sorcin target in cardiac muscle. In this study, RNAi knockdown of sorcin, purified sorcin or sorcin variants was employed in parallel measurements of: (i) NCX activity in isolated rabbit cardiomyocytes using electrophysiological techniques and (ii) sorcin binding to the NCX1 calcium binding domains (CBD1 and (iii) using surface plasmon resonance and gel overlay techniques. Sorcin is activated by Ca2+ binding to the EF3 and EF2 regions, which are connected by the D helix. To investigate the importance of this region in the interaction with NCX1, three variants were examined: W105G and W99G, mutated respectively near EF3 and EF2, and E124A that does not bind Ca2+ due to a mutation at EF3. Downregulation of sorcin decreased and supplementation with wt sorcin (3 μM) increased NCX activity in isolated cardiomyocytes. The relative stimulatory effects of the sorcin variants were: W105G > wt sorcin > Sorcin Calcium Binding Domain (SCBD) > W99G > E124A. Sorcin binding to both CBD1 and 2 was observed. In the presence of 50 µM Ca2+, the interaction with CBD1 followed the order W105G > SCBD > wt sorcin > W99G > E124A. In sorcin, the interacting surface can be mapped on the C-terminal Ca2+-binding domain in the D helix region comprising W99. The fast association/dissociation rates that characterize the interaction of sorcin with CBD1 and 2 may permit complex formation/dissociation during an excitation/contraction cycle.


Biochimica et Biophysica Acta | 2014

The Salmonella enterica ZinT structure, zinc affinity and interaction with the high-affinity uptake protein ZnuA provide insight into the management of periplasmic zinc

Andrea Ilari; Flaminia Alaleona; Giancarlo Tria; Patrizia Petrarca; Andrea Battistoni; Carlotta Zamparelli; Daniela Verzili; Mattia Falconi; Emilia Chiancone

BACKGROUND In Gram-negative bacteria the ZnuABC transporter ensures adequate zinc import in Zn(II)-poor environments, like those encountered by pathogens within the infected host. Recently, the metal-binding protein ZinT was suggested to operate as an accessory component of ZnuABC in periplasmic zinc recruitment. Since ZinT is known to form a ZinT-ZnuA complex in the presence of Zn(II) it was proposed to transfer Zn(II) to ZnuA. The present work was undertaken to test this claim. METHODS ZinT and its structural relationship with ZnuA have been characterized by multiple biophysical techniques (X-ray crystallography, SAXS, analytical ultracentrifugation, fluorescence spectroscopy). RESULTS The metal-free and metal-bound crystal structures of Salmonella enterica ZinT show one Zn(II) binding site and limited structural changes upon metal removal. Spectroscopic titrations with Zn(II) yield a KD value of 22±2nM for ZinT, while those with ZnuA point to one high affinity (KD<20nM) and one low affinity Zn(II) binding site (KD in the micromolar range). Sedimentation velocity experiments established that Zn(II)-bound ZinT interacts with ZnuA, whereas apo-ZinT does not. The model of the ZinT-ZnuA complex derived from small angle X-ray scattering experiments points to a disposition that favors metal transfer as the metal binding cavities of the two proteins face each other. CONCLUSIONS ZinT acts as a Zn(II)-buffering protein that delivers Zn(II) to ZnuA. GENERAL SIGNIFICANCE Knowledge of the ZinT-ZnuA relationship is crucial for understanding bacterial Zn(II) uptake.


The International Journal of Biochemistry & Cell Biology | 2012

The N-terminal extension is essential for the formation of the active dimeric structure of liver peroxisomal alanine:glyoxylate aminotransferase.

Riccardo Montioli; Sonia Fargue; Jackie Lewin; Carlotta Zamparelli; Christopher J. Danpure; Carla Borri Voltattorni; Barbara Cellini

Alanine:glyoxylate aminotransferase (AGT) is a pyridoxal-phosphate (PLP)-dependent enzyme. Its deficiency causes the hereditary kidney stone disease primary hyperoxaluria type 1. AGT is a highly stable compact dimer and the first 21 residues of each subunit form an extension which wraps over the surface of the neighboring subunit. Naturally occurring and artificial amino acid replacements in this extension create changes in the functional properties of AGT in mammalian cells, including relocation of the enzyme from peroxisomes to mitochondria. In order to elucidate the structural and functional role of this N-terminal extension, we have analyzed the consequences of its removal using a variety of biochemical and cell biological methods. When expressed in Escherichia coli, the N-terminal deleted form of AGT showed the presence of the protein but in an insoluble form resulting in only a 10% soluble yield as compared to the full-length version. The purified soluble fraction showed reduced affinity for PLP and greatly reduced catalytic activity. Although maintaining a dimer form, it was highly prone to self-aggregation. When expressed in a mammalian cell line, the truncated construct was normally targeted to peroxisomes, where it formed large stable but catalytically inactive aggregates. These results suggest that the N-terminal extension plays an essential role in allowing AGT to attain its correct conformation and functional activity. The precise mechanism of this effect is still under investigation.

Collaboration


Dive into the Carlotta Zamparelli's collaboration.

Top Co-Authors

Avatar

Emilia Chiancone

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Daniela Verzili

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Andrea Ilari

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Gianni Colotti

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Alberto Boffi

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge