Network


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

Hotspot


Dive into the research topics where Alessandra Dorcaratto is active.

Publication


Featured researches published by Alessandra Dorcaratto.


Neurochemistry International | 2006

Expression of CXC chemokine receptors 1-5 and their ligands in human glioma tissues : Role of CXCR4 and SDF1 in glioma cell proliferation and migration

Adriana Bajetto; Federica Barbieri; Alessandra Dorcaratto; Simone Barbero; Antonio Daga; Carola Porcile; Jean Louis Ravetti; Gianluigi Zona; Renato Spaziante; Giorgio Corte; Gennaro Schettini; Tullio Florio

Chemokines have been involved in cellular processes associated to malignant transformation such as proliferation, migration and angiogenesis. The expression of five CXC chemokine receptors and their main ligands was analysed by RT-PCR in 31 human astrocytic neoplasms. The mRNAs for all the receptors analysed were identified in a high percentage of tumours, while their ligands showed lower expression. CXCR4 and SDF1 were the most frequently mRNA identified (29/31 and 13/31 of the gliomas studied, respectively). Thus, we further analysed the cell localization of CXCR4 and SDF1 in immunohistochemistry experiments. We show a marked co-localization of CXCR4 and SDF1 in tumour cells, mainly evident in psudolpalisade and microcystic degeneration areas and in the vascular endothelium. In addition, hSDF1alpha induced a significant increase of DNA synthesis in primary human glioblastoma cell cultures and chemotaxis in a glioblastoma cell line. These results provide evidence of the expression of multiple CXC chemokines and their receptors in brain tumours and that in particular CXCR4 and SDF1 sustain proliferation and migration of glioma cells to promote malignant progression.


American Journal of Pathology | 2002

Differentiation between high- and low-grade astrocytoma using a human recombinant antibody to the extra domain-B of fibronectin.

Patrizia Castellani; Laura Borsi; Barbara Carnemolla; Attila Birò; Alessandra Dorcaratto; Giuseppe L. Viale; Dario Neri; Luciano Zardi

Different fibronectin (FN) isoforms are generated by the alternative splicing of the primary FN transcript. We previously demonstrated that the isoform containing the extra domain B sequence of fibronectin (B-FN), a complete type-III-homology repeat, is a marker of angiogenesis that accumulates around neovasculature only during angiogenic processes. We produced a single-chain human recombinant antibody (scFv), L19, which reacts specifically with B-FN and selectively targets tumor vasculature in vivo. We used this scFv and an antibody against a pan-endothelial marker (Factor VIII) in a double-staining procedure on specimens of low- and high-grade astrocytomas to determine the percentage of B-FN-positive vessels, (denominating the resulting value angiogenic index [AI]). Compared to vascular density and proliferative activity (evaluated using antibodies to Factor VIII and Ki67, respectively), AI correlated better with tumor grade (1.6 +/- 2.6% and 92.0 +/- 8.7% of B-FN-positive vessels in low- and high-grade astrocytomas, respectively) and was a more precise diagnostic tool than either of the two conventional methods. In fact, discriminating analysis using these three parameters showed that only AI accurately classified 100% of the cases studied, compared to 64% and 89% correctly diagnosed by vascular density and of proliferating cells, respectively.


American Journal of Pathology | 1999

Identification of a Glioblastoma-Associated Tenascin-C Isoform by a High Affinity Recombinant Antibody

Barbara Carnemolla; Patrizia Castellani; Marco Ponassi; Laura Borsi; Stefania Urbini; Guido Nicolò; Alessandra Dorcaratto; Giuseppe L. Viale; Greg Winter; Dario Neri; Luciano Zardi

Tenascin-C exists in several polymorphic isoforms due to alternative splicing of nine fibronectin-like type III repeats. Large Tenascin-C isoforms are present in almost all normal adult tissues but are upregulated in fetal, regenerating, and neoplastic tissues. Here, we report a human antibody fragment, TN11, derived from a phage library with high affinity for the spliced repeat C and demonstrate that this repeat is undetectable in normal adult tissues, barely detectable or undetectable in breast, lung and gastric carcinomas, meningioma, and low grade astrocytoma, but extremely abundant in high grade astrocytoma (grade III and glioblastoma), especially around vascular structures and proliferating cells. The antibody appears to have potential for development of a therapeutic agent for patients with high grade astrocytoma.


Clinical Cancer Research | 2008

Overexpression of Stromal Cell–Derived Factor 1 and Its Receptor CXCR4 Induces Autocrine/Paracrine Cell Proliferation in Human Pituitary Adenomas

Federica Barbieri; Adriana Bajetto; Ralf Stumm; Alessandra Pattarozzi; Carola Porcile; Gianluigi Zona; Alessandra Dorcaratto; Jean Louis Ravetti; Francesco Minuto; Renato Spaziante; Gennaro Schettini; Diego Ferone; Tullio Florio

Purpose: Hypothalamic or locally produced growth factors and cytokines control pituitary development, functioning, and cell division. We evaluated the expression of the chemokine stromal cell–derived factor 1 (SDF1) and its receptor CXCR4 in human pituitary adenomas and normal pituitary tissues and their role in cell proliferation. Experimental Design: The expression of SDF1 and CXCR4 in 65 human pituitary adenomas and 4 human normal pituitaries was determined by reverse transcription-PCR, immunohistochemistry, and confocal immunofluorescence. The proliferative effect of SDF1 was evaluated in eight fibroblast-free human pituitary adenoma cell cultures. Results: CXCR4 mRNA was expressed in 92% of growth hormone (GH)-secreting pituitary adenomas (GHoma) and 81% of nonfunctioning pituitary adenomas (NFPA), whereas SDF1 was identified in 63% and 78% of GHomas and NFPAs, respectively. Immunostaining for CXCR4 and SDF1 showed a strong homogenous labeling in all tumoral cells in both GHomas and NFPAs. In normal tissues, CXCR4 and SDF1 were expressed only in a subset of anterior pituitary cells, with a lower expression of SDF1 compared with its cognate receptor. CXCR4 and SDF1 were not confined to a specific cell population in the anterior pituitary but colocalized with discrete subpopulations of GH-, prolactin-, and adrenocorticorticotropic hormone–secreting cells. Conversely, most of the SDF1-containing cells expressed CXCR4. In six of eight pituitary adenoma primary cultures, SDF1 induced a statistically significant increase in DNA synthesis that was prevented by the treatment with the CXCR4 antagonist AMD3100 or somatostatin. Conclusions: CXCR4 and SDF1 are overexpressed in human pituitary adenomas and CXCR4 activation may contribute to pituitary cell proliferation and, possibly, to adenoma development `in humans.


Cancer | 1997

Tumor targeting potential of the monoclonal antibody BC‐1 against oncofetal fibronectin in nude mice bearing human tumor implants

Giuliano Mariani; Arben Lasku; Enrica Balza; Barbara Gaggero; Cinzia Motta; Lorella Di Luca; Alessandra Dorcaratto; Giuseppe A. Viale; Dario Neri; Luciano Zardi

The immunoglobulin G1 (IgG1) monoclonal antibody (MoAb) BC‐1 detects human oncofetal fibronectin, which has extremely restricted distribution in normal adult tissues and is highly expressed in fetal and tumor tissues.


Journal of Neuro-oncology | 2004

Expression of Somatostatin Receptor mRNA in Human Meningiomas and their Implication in in vitro Antiproliferative Activity

Sara Arena; Federica Barbieri; Stefano Thellung; Paolo Pirani; Alessandro Corsaro; Valentina Villa; Patrizia Dadati; Alessandra Dorcaratto; Gabriella Lapertosa; Jean-Louis Ravetti; Renato Spaziante; Gennaro Schettini; Tullio Florio

Somatostatin receptors (SSTRs) have been detected in many normal and malignant tissues. This wide expression has been used for diagnostic, prognostic and therapeutic purposes. Five SSTR subtypes (SSTR 1–5) have been identified whose activation is responsible for the signal transduction through many different intracellular pathways. In the present study the expression of SSTR mRNA was determined by reverse-transcriptase (RT)–PCR in 42 meningiomas. About 88% of the tumors analyzed (37/42) were positive for at least one of the five SSTR subtypes displaying a variable pattern of expression of the different SSTR subtypes. SSTR1 and SSTR2 were the most frequently mRNA detected (69% and 79% of the sample analyzed, respectively). The other subtypes were found in the 43%, 33% and 33% of cases for SSTR3, SSTR4 and SSTR5, respectively. In 22, out of 42 patients (52%) three or more SSTRs were detected. The expression of the different SSTR subtypes did not correlate with the expression of bcl-2 (apoptosis-associated protein) and MIB-1 (a proliferation marker), assessed by immunohistochemistry in a series of 34 tumor samples, while a correlation between the expression of SSTR3 and p53 was observed (p = 0.08). To evaluate a possible role of SSTR in the control of human meningioma cell proliferation, seven primary cell cultures obtained from fresh meningioma surgical tissues, were analyzed for their proliferative behavior by MTT assay and for their response to SST by [3H]-thymidine incorporation. In four out of six tumors (in one case no SSTR were detected) the treatment with SST caused a significant inhibition of DNA synthesis induced by the tumor-promoter phorbol myristate acetate. The evidence of the expression of SSTRs, mainly of SSTR2, in this series of specimens we analyzed altogether with in vitro antiproliferative effects of SST may open interesting perspectives for the diagnosis and the therapy of meningiomas.


Neuro-oncology | 2007

CXCR4 and SDF1 expression in human meningiomas: A proliferative role in tumoral meningothelial cells in vitro

Adriana Bajetto; Federica Barbieri; Alessandra Pattarozzi; Alessandra Dorcaratto; Carola Porcile; Jean Louis Ravetti; Gianluigi Zona; Renato Spaziante; Gennaro Schettini; Tullio Florio

Chemokines participate in cellular processes associated with tumor proliferation, migration, and angiogenesis. We previously demonstrated that stromal cell-derived factor 1 (SDF1) exerts a mitogenic activity in glioblastomas through the activation of its receptor CXCR4. Here we studied the expression of this chemokine in human meningiomas and its possible role in cell proliferation. Reverse transcriptase-PCR analysis for CXCR4 and SDF1 was performed on 55 human meningiomas (47 WHO grade I, 5 WHO II, and 3 WHO III). Immunolabeling for CXCR4 and SDF1 was performed on paraffin-embedded sections of these tumors. [(3)H]Thymidine uptake and Western blot analyses were performed on primary meningeal cell cultures of tumors to evaluate the proliferative activity of human SDF1alpha (hSDF1alpha) in vitro and the involvement of extracellular signal-regulated kinase 1/2 (ERK1/2) activation in this process. CXCR4 mRNA was expressed by 78% of the tumor specimens and SDF1 mRNA by 53%. CXCR4 and SDF1 were often detected in the same tumor tissues and colocalized with epithelial membrane antigen immunostaining. In 9 of 12 primary cultures from meningiomas, hSDF1alpha induced significant cell proliferation that was strongly reduced by the mitogen-activated protein kinase kinase inhibitor PD98059, involving ERK1/2 activation in the proliferative signal of hSDF1alpha. In fact, CXCR4 stimulation led to ERK1/2 phosphorylation/activation. In addition, the hSDF1alpha-induced cell proliferation was significantly correlated with the MIB1 staining index in the corresponding surgical specimen. In conclusion, we found that human meningiomas express CXCR4 and SDF1 and that hSDF1alpha induces proliferation in primary meningioma cell cultures through the activation of ERK1/2.


Annals of the New York Academy of Sciences | 2006

CXC receptor and chemokine expression in human meningioma: SDF1/CXCR4 signaling activates ERK1/2 and stimulates meningioma cell proliferation

Federica Barbieri; Adriana Bajetto; Carola Porcile; Alessandra Pattarozzi; Alessandro Massa; Gianluigi Lunardi; Gianluigi Zona; Alessandra Dorcaratto; Jean Louis Ravetti; Renato Spaziante; Gennaro Schettini; Tullio Florio

Abstract:  Recent evidence indicates that cancer cells express chemokine (CK) receptors and that their signaling is crucial for tumor proliferation, migration, and angiogenesis. The profiles of expression of CXC CK receptors (CXCR1‐5) and their main ligands (growth‐related oncogene, GRO1‐2‐3/CXCL1‐2‐3; interleukin 8, IL‐8/CXCL8; monokine‐induced γ‐interferon MIG/CXCL9; γ‐interferon‐inducible‐protein‐10, IP‐10/CXCL10; stromal cell‐derived factor‐1, SDF1/CXCL12; B‐cell activating CK‐1, BCA‐1/CXCL13) were analyzed by reverse transcription polymerase chain reaction (RT‐PCR) in surgical samples of human meningiomas. All the five receptors displayed high percentages of positive cases: 92% CXCR1, 89% CXCR2, 83% CXCR3, 78% CXCR4, and 94% CXCR5. Conversely, their ligands showed a lower pattern of expression: 40% IL‐8, 42% GRO1‐3, 42% IP‐10, 28% MIG, 53% SDF1, and 3% BCA‐1. SDF1/CXCR4 interaction plays a pivotal role in cancer proliferation. Thus, the signaling mechanisms activated by the exclusive binding between SDF1 and CXCR4 was investigated in 12 primary cultures from meningioma tissues. CXCR4 was functionally coupled as demonstrated by the significant increase of DNA synthesis in meningioma cells in response to SDF1, measured by [3H]‐thymidine uptake. In three primary cultures, the SDF1‐dependent mitogenic activity was associated with a marked phosphorylation of extracellular signal‐regulated kinase (ERK1/2) as evaluated by Western blots. PD98059 (a MEK inhibitor) significantly reduced ERK1/2 activation, thus linking the SDF1/CXCR4 pathway to meningioma cell proliferation via ERK1/2 signal transduction. We demonstrate, for the first time in human meningiomas, the simultaneous expression of CXCR1‐5 and their CKs and the mitogenic activity of SDF1/CXCR4, suggesting a pivotal role of these receptor–ligand pairs in meningeal tumors.


Cancer | 1997

A pilot pharmacokinetic and immunoscintigraphic study with the technetium-99m-labeled monoclonal antibody BC-1 directed against oncofetal fibronectin in patients with brain tumors.

Giuliano Mariani; Arben Lasku; Antonio Pau; Giuseppe Villa; Cinzia Motta; Giuseppina Calcagno; Gioconda Z. Taddei; Patrizia Castellani; Kostas Syrigos; Alessandra Dorcaratto; Agamennon A. Epenetos; Luciano Zardi; Giuseppe A. Viale

Preliminary experiments in an animal model have shown the favorable tumor targeting potential in vivo of radiolabeled BC‐1, an immunoglobulin (Ig)G1 monoclonal antibody (MoAb) that recognizes the human fibronectin isoform (B+) containing the ED‐B oncofetal domain. This antigen has extremely restricted distribution in normal adult tissues. Instead, it is highly expressed in fetal and tumor tissues, especially in high grade astrocytomas and malignant gliomas of the brain, in which the process of neoangiogenesis linked to tumor growth is particularly important.


Acta Neurochirurgica | 1995

Tenascin distribution in human brain tumours

Patrizia Inst.Naz.Ric Castellani; Alessandra Dorcaratto; Annalisa Siri; Luciano Zardi; Giuseppe L. Viale

SummaryUsing a monoclonal antibody specific for human tenascin (TN), 180 intracranial growths were immunohistochemically studied.In 69 cases of meningioma, neoplastic cells were negative, with some positivity being observed only in the perivascular and the supporting stroma, especially in anaplastic meningiomas. In 57 cases of glioma different degrees of reactivity occurred in both the cellular conglomerates and the stromal components of the tumours. A higher variability in reactivity was observed in anaplastic astrocytomas and glioblastomas.The most constant finding of the study was the staining of the stroma, which was observed in all types of growths, including metastasis, abscess and tuberculoma.The results are consistent with the hypothesis that tenascin is a stromal marker rather than a true marker of malignant tumours.The heterogeneous distribution of TN in anaplastic gliomas may be a factor in the variable response to treatment with radiolabelled anti-TN monoclonal antibodies.

Collaboration


Dive into the Alessandra Dorcaratto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luciano Zardi

Istituto Giannina Gaslini

View shared research outputs
Top Co-Authors

Avatar

Patrizia Castellani

National Cancer Research Institute

View shared research outputs
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