Network


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

Hotspot


Dive into the research topics where Antonio Melcarne is active.

Publication


Featured researches published by Antonio Melcarne.


Human Molecular Genetics | 2010

The genetic and metabolic signature of oncocytic transformation implicates HIF1α destabilization

Anna Maria Porcelli; Anna Ghelli; Claudio Ceccarelli; Martin Lang; Giovanna Cenacchi; Mariantonietta Capristo; Lucia Fiammetta Pennisi; Isabella Morra; Enrica Ciccarelli; Antonio Melcarne; Anna Bartoletti-Stella; Nunzio Salfi; Giovanni Tallini; Andrea Martinuzzi; Valerio Carelli; Marcella Attimonelli; Michela Rugolo; Giovanni Romeo; Giuseppe Gasparre

We previously showed that disruptive complex I mutations in mitochondrial DNA are the main genetic hallmark of oncocytic tumors of the thyroid and kidney. We here report a high frequency of homoplasmic disruptive mutations in a large panel of oncocytic pituitary and head-and-neck tumors. The presence of such mutations implicates disassembly of respiratory complex I in vivo which in turn contributes to the inability of oncocytic tumors to stabilize HIF1alpha and to display pseudo-hypoxia. By utilizing transmitochondrial cytoplasmic hybrids (cybrids), we induced the shift to homoplasmy of a truncating mutation in the mitochondria-coded MTND1 gene. Such shift is associated with a profound metabolic impairment leading to the imbalance of alpha-ketoglutarate and succinate, the Krebs cycle metabolites which are the main responsible for HIF1alpha stabilization. We conclude that the main hallmarks of oncocytic transformation, namely the occurrence of homoplasmic disruptive mutations and complex I disassembly, may explain the benign nature of oncocytic neoplasms through lack of HIF1alpha stabilization.


Cell Biochemistry and Function | 2010

Glioblastoma cancer stem cells: heterogeneity, microenvironment and related therapeutic strategies

Tetyana Denysenko; Luisa Gennero; Maria Augusta Roos; Antonio Melcarne; Carola Juenemann; Giuliano Faccani; Isabella Morra; G. P. Cavallo; Stefano Reguzzi; Gianpiero Pescarmona; Antonio Ponzetto

Glioblastoma Multiforme (GBM) is an incurable malignancy. GBM patients have a short life expectancy despite aggressive therapeutic approaches based on surgical resection followed by adjuvant radiotherapy and concomitant chemotherapy. Glioblastoma growth is characterized by a high motility of tumour cells, their resistance to both chemo/radio‐therapy, apoptosis inhibition leading to failure of conventional therapy. Cancer Stem Cells (CSCs), identified in GBM as well as in many other cancer types, express the membrane antigen prominin‐1 (namely CD133). These cells and normal Neural Stem Cells (NSC) share surface markers and properties, i.e. are able to self‐renew and differentiate into multiple cell types. Stem cell self‐renewal depends on microenvironmental cues, including Extracellular Matrix (ECM) composition and cell types. Therefore, the role of microenvironment needs to be evaluated to clarify its importance in tumour initiation and progression through CSCs. The specific microenvironment of CSCs was found to mimic in part the vascular niche of normal stem cells. The targeting of GMB CSCs may represent a powerful treatment approach. Lastly, in GBM patients cancer‐initiating cells contribute to the profound immune suppression that in turn correlated with CSCs STAT3 (CD133 + ). Further studies of microenvironment are needed to better understand the origin of GMB/GBM CSCs and its immunosuppressive properties. Copyright


BioMed Research International | 2014

Stem Cell Niches in Glioblastoma: A Neuropathological View

Davide Schiffer; Marta Mellai; Laura Annovazzi; Valentina Caldera; Angela Piazzi; Tetyana Denysenko; Antonio Melcarne

Glioblastoma (GBM) stem cells (GSCs), responsible for tumor growth, recurrence, and resistance to therapies, are considered the real therapeutic target, if they had no molecular mechanisms of resistance, in comparison with the mass of more differentiated cells which are insensitive to therapies just because of being differentiated and nonproliferating. GSCs occur in tumor niches where both stemness status and angiogenesis are conditioned by the microenvironment. In both perivascular and perinecrotic niches, hypoxia plays a fundamental role. Fifteen glioblastomas have been studied by immunohistochemistry and immunofluorescence for stemness and differentiation antigens. It has been found that circumscribed necroses develop inside hyperproliferating areas that are characterized by high expression of stemness antigens. Necrosis developed inside them because of the imbalance between the proliferation of tumor cells and endothelial cells; it reduces the number of GSCs to a thin ring around the former hyperproliferating area. The perinecrotic GSCs are nothing else that the survivors remnants of those populating hyperproliferating areas. In the tumor, GSCs coincide with malignant areas so that the need to detect where they are located is not so urgent.


International Journal of Oncology | 2015

The DNA damage/repair cascade in glioblastoma cell lines after chemotherapeutic agent treatment

Laura Annovazzi; Valentina Caldera; Marta Mellai; Chiara Riganti; Luigi Battaglia; Daniela Chirio; Antonio Melcarne; Davide Schiffer

Therapeutic resistance in glioblastoma multiforme (GBM) has been linked to a subpopulation of cells with stem cell-like properties, the glioma stem cells (GSCs), responsible for cancer progression and recurrence. This study investigated the in vitro cytotoxicity of three chemotherapeutics, temozolomide (TMZ), doxorubicin (Dox) and paclitaxel (PTX) on glioma cell lines, by analyzing the molecular mechanisms leading to DNA repair and cell resistance, or to cell death. The drugs were tested on 16 GBM cell lines, grown as neurospheres (NS) or adherent cells (AC), by studying DNA damage occurrence by Comet assay, the expression by immunofluorescence and western blotting of checkpoint/repair molecules and apoptosis. The three drugs were able to provoke a genotoxic injury and to inhibit dose- and time-dependently cell proliferation, more evidently in AC than in NS. The first cell response to DNA damage was the activation of the damage sensors (p-ATM, p-53BP1, γ-H2AX), followed by repair effectors; the expression of checkpoint/repair molecules appeared higher in NS than in AC. The non-homologous repair pathway (NHEJ) seemed more involved than the homologous one (HR). Apoptosis occurred after long treatment times, but only a small percentage of cells in NS underwent death, even at high drug concentration, whereas most cells survived in a quiescent state and resumed proliferation after drug removal. In tumor specimens, checkpoint/repair proteins were constitutively expressed in GBMs, but not in low-grade gliomas.


Cell Biochemistry and Function | 2013

Protective effects of polydeoxyribonucleotides on cartilage degradation in experimental cultures

Luisa Gennero; Tetyana Denysenko; Gian Franco Calisti; Andrea Vercelli; Carlo Vercelli; S. Amedeo; Silvia Mioletti; Enrico Parino; Manuela Gessica Montanaro; Antonio Melcarne; Carola Juenemann; Enrico De Vivo; Alessandro Longo; G. P. Cavallo; Rocco De Siena

The capacity of cartilage self‐regeneration is considered to be limited. Joint injuries often evolve in the development of chronic wounds on the cartilage surface. Such lesions are associated with articular cartilage degeneration and osteoarthritis. Re‐establishing a correct micro/macro‐environment into damaged joints could stop or prevent the degenerative processes. This study investigated the effect of polydeoxyribonucleotides (PDRNs) on cartilage degradation in vitro and on cartilage extracted cells. The activities of matrix metalloproteinases 2 and 9 were measured in PDRN‐treated cells and in controls at days 0 and 30 of culture. Human nasal cartilage explants were cultured, and the degree of proteoglycan degradation was assessed by measuring the amount of glycosaminoglycans released into the culture medium. The PDRN properties compared with controls were tested on cartilage tissues to evaluate deposition of extracellular matrix. Chondrocytes treated with PDRNs showed a physiological deposition of extracellular matrix (aggrecan and type II collagen: Western blot, IFA, fluorescence activated cell sorting, Alcian blue and safranin O staining). PDRNs were able to inhibit proteoglycan degradation in cartilage explants. The activities of matrix metalloproteinases 2 and 9 were reduced in all PDRN‐treated samples. Our results indicate that PDRNs are suitable for a long‐term cultivation of in vitro cartilage and have therapeutic effects on chondrocytes by protecting cartilage. Copyright


OncoTargets and Therapy | 2015

Astroblastoma: beside being a tumor entity, an occasional phenotype of astrocytic gliomas?

Marta Mellai; Angela Piazzi; Cristina Casalone; Silvia Grifoni; Antonio Melcarne; Laura Annovazzi; Paola Cassoni; Tetyana Denysenko; Maria Consuelo Valentini; Angelina Cistaro; Davide Schiffer

The diagnosis of astroblastoma is based on a typical histological aspect with perivascular distribution of cells sending cytoplasmic extensions to the vessels and vascular hyalinization. These criteria are useful for standardizing the identification of the tumor, but, in spite of this, there are discrepancies in the literature concerning the age distribution and the benign or malignant nature of the tumor. Three cases are discussed in this study: Case 1 was a typical high-grade astroblastoma; Case 2 was an oligodendroglioma at the first intervention and an oligoastrocytoma at the second intervention with typical perivascular arrangements in the astrocytic component; Case 3 was a gemistocytic glioma with malignant features and typical perivascular arrangements. Genetic analysis showed genetic alterations that are typical of gliomas of all malignancy grades. Using the neurosphere assay, neurospheres and adherent cells were found to have developed in Case 1, while adherent cells only developed in Case 2, in line with the stemness potential of the tumors. The cases are discussed in relation to their diagnostic assessment as astroblastoma, and it is hypothesized that the typical perivascular distribution of cells may not indicate a separate and unique tumor entity, but may be a peculiarity that can be acquired by astrocytic gliomas when an unknown cause from the tumor microenvironment influences the relationship between vessels and tumor cells.


Cell Biochemistry and Function | 2014

Heterogeneous phenotype of human glioblastoma: In vitro study

Tetyana Denysenko; Luisa Gennero; Carola Juenemann; Isabella Morra; Paolo Masperi; Vincenzo Ceroni; Antonella Pragliola; Antonio Ponzetto; Antonio Melcarne

Glioblastomas (GBMs) are the most lethal primary brain tumours. Increasing evidence shows that brain tumours contain the population of stem cells, so‐called cancer stem cells (CSCs). Stem cell marker CD133 was reported to identify CSC population in GBM. Further studies have indicated that CD133 negative cells exhibiting similar properties and are able to initiate the tumour, self‐renew and undergo multilineage differentiation. GBM is a highly heterogeneous tumour and may contain different stem cell populations with different functional properties. We characterized five GBM cell lines, established from surgical samples, according to the marker expression, proliferation and differentiation potential. CD133 positive cell lines showed increased proliferation rate in neurosphere condition and marked differentiation potential towards neuronal lineages. Whereas two cell lines low‐expressing CD133 marker showed mesenchymal properties in vitro, that is high proliferation rate in serum condition and differentiation in mesenchymal cell types. Further, we compared therapy resistance capacity of GBM cell lines treated with hydroxyurea. Our results suggest that CSC concept is more complex than it was believed before, and CD133 could not define entire stem cell population within GBM. At least two different subtypes of GBM CSCs exist, which may have different biological characteristics and imply different therapeutic strategies. Copyright


Oncotarget | 2017

Comparison among conventional and advanced MRI, 18 F-FDG PET/CT, phenotype and genotype in glioblastoma

Maria Consuelo Valentini; Marta Mellai; Laura Annovazzi; Antonio Melcarne; Tetyana Denysenko; Paola Cassoni; Cristina Casalone; Cristiana Maurella; Silvia Grifoni; Piercarlo Fania; Angelina Cistaro; Davide Schiffer

Glioblastoma (GB) is a highly heterogeneous tumor. In order to identify in vivo the most malignant tumor areas, the extent of tumor infiltration and the sites giving origin to GB stem cells (GSCs), we combined positron emission tomography/computed tomography (PET/CT) and conventional and advanced magnetic resonance imaging (MRI) with histology, immunohistochemistry and molecular genetics. Prior to dura opening and tumor resection, forty-eight biopsy specimens [23 of contrast-enhancing (CE) and 25 of non-contrast enhancing (NE) regions] from 12 GB patients were obtained by a frameless image-guided stereotactic biopsy technique. The highest values of 2-[18F]-fluoro-2-deoxy-D-glucose maximum standardized uptake value (18F-FDG SUVmax), relative cerebral blood volume (rCBV), Choline/Creatine (Cho/Cr), Choline/N-acetylaspartate (Cho/NAA) and Lipids/Lactate (LL) ratio have been observed in the CE region. They corresponded to the most malignant tumor phenotype, to the greatest molecular spectrum and stem cell potential. On the contrary, apparent diffusion coefficient (ADC) and fractional anisotropy (FA) in the CE region were very variable. 18F-FDG SUVmax, Cho/Cr and Cho/NAA ratio resulted the most suitable parameters to detect tumor infiltration. In edematous areas, reactive astrocytes and microglia/macrophages were influencing variables. Combined MRI and 18F-FDG PET/CT allowed to recognize the specific biological significance of the different identified areas of GB.


Oncotarget | 2018

Correction: SEL1L SNP rs12435998, a predictor of glioblastoma survival and response to radio-chemotherapy

Marta Mellai; Monica Cattaneo; Alessandra Maria Storaci; Laura Annovazzi; Paola Cassoni; Antonio Melcarne; Pasquale De Blasio; Davide Schiffer; Ida Biunno

[This corrects the article DOI: 10.18632/oncotarget.3611.].


Cancer Genomics & Proteomics | 2016

WNT/ β- catenin Signaling Pathway and Downstream Modulators in Low- and High-grade Glioma

Tetyana Denysenko; Laura Annovazzi; Paola Cassoni; Antonio Melcarne; Marta Mellai; Davide Schiffer

Collaboration


Dive into the Antonio Melcarne's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge