129 research outputs found

    Strategies for an urban renewal in Rome: Massimina Co_Goal

    Get PDF
    Abstract The paper presents a research project about the regeneration of an informal neighbourhood in Rome. To address the complexity of the regeneration process, the project relies on a combined top down/bottom up strategy. The project envisions the implementation of a Public Call to promote a set of retrofitting actions for a defined number of private houses to be involved in a co-financed refurbishment program. Main goal of the Public Call is to foster the transition towards sustainable development, transforming the existing district into a low energy district and developing new community services implemented and managed by a local community company

    Expression of angiogenesis related factors in glioblastoma and peritumor tissue

    Get PDF
    Glioblastoma (GBM) is a lethal brain glial tumor characterized by extensive angiogenesis that is mostly triggered by tumor hypoxia. We previous reported that in GBM and in peritumor areas, endothelial cells expressed CD105, which probably marks newly formed vessels with a quite similar morphology. In this study, with the aim of better understanding the involvement of angiogenesis in tumor progression, we analyzed, by immunohistochemistry, the expression of Hypoxia-inducible factor (HIF) 1α and 2α, VEGF and its receptors (VEGFR-1 and -2) in GBM and in peritumor tissue. Twenty two patients were enrolled in this study. Tissue specimens were derived from enhanced lesion (first area) and white matter at a distance ≤1 mm from the tumor edge (second area). Immunoreactivity for all markers was detected not only in the tumor but also in the peritumor tissue and it was present in neoplastic cells, in endothelium and in apparently normal glial cells. HIF1α and 2α expression was mainly confined in the nuclei. VEGF, localized in the cytoplasm, showed diffuse expression with an intense staining in GBM. VEGFR-1 and 2 immupositivity was localized especially to the cell membrane and also to the cytoplasm, as expected. All molecule staining was evident in a heterogeneous manner and there was no significant difference in the expression marker levels between the first and second area also in relation to the presence or absence of tumor cells in the second area. No significant correlation was found between the above molecule expression and survival time. In conclusion, we demonstrated that HIF1α, HIF2α, VEGF and VEGFR-1 and -2 are present in peritumor area, probably reflecting perturbations of oxygenation emanating from the tumor microenvironment. Since, unfortunately, the response to anti-VEGF therapy is transient and the majority of patients eventually relapse, the gain of a deeper knowledge of the above molecule role, in the peritumor tissue, may lead to consider them as the target for new treatment regimens to counteract angiogenesis. Supported by FIRB “Accordi di Programma” 201

    PDGFRα depletion attenuates glioblastoma stem cells features by modulation of STAT3, RB1 and multiple oncogenic signals.

    Get PDF
    Platelet derived growth factor receptors (PDGFRs) play an important role in tumor pathogenesis, and they are frequently overexpressed in glioblastoma (GBM). Earlier we have shown a higher protein expression of PDGFR isoforms (α and β) in peritumoral-tissue derived cancer stem cells (p-CSC) than in tumor core (c-CSC) of several GBM affected patients. In the current study, in order to assess the activity of PDGFRα/PDGF-AA signaling axis, we performed time course experiments to monitor the effects of exogenous PDGF-AA on the expression of downstream target genes in c-CSC vs p-CSC. Interestingly, in p-CSC we detected the upregulation of Y705-phosphorylated Stat3, concurrent with a decrement of Rb1 protein in its active state, within minutes of PDGF-AA addition. This finding prompted us to elucidate the role of PDGFRα in self-renewal, invasion and differentiation in p-CSC by using short hairpin RNA depletion of PDGFRα expression. Notably, in PDGFRα-depleted cells, protein analysis revealed attenuation of stemness-related and glial markers expression, alongside early activation of the neuronal marker MAP2a/b that correlated with the induction of tumor suppressor Rb1. The in vitro reduction of the invasive capacity of PDGFRα-depleted CSC as compared to parental cells correlated with the downmodulation of markers of epithelial-mesenchymal transition phenotype and angiogenesis. Surprisingly, we observed the induction of anti-apoptotic proteins and compensatory oncogenic signals such as EDN1, EDNRB, PRKCB1, PDGF-C and PDGF-D. To conclude, we hypothesize that the newly discovered PDGFRα/Stat3/Rb1 regulatory axis might represent a potential therapeutic target for GBM treatment.BRC, Q

    Relevance of tumour surrounding area in chemoresistance of glioblastoma (GBM)

    Get PDF
    The mechanisms responsible for resistance to damage in normal cells might determine chemoresistance in both tumor cells and cancer stem cells (CSC). Relapse due to chemoresistant residual disease is a major cause of death in GBM. Increasing body of evidence indicates that not only tumor area (TT), but also tissue surrounding the tumor border (pTT) of GBM contains tumor cells and CSC, which could contribute to the disease progression. Therefore, the need to have a deeper insight in this area through identification of the characteristics that confer chemoresistance. In this study, the expression of molecules involved in chemoresistance was investigated in samples derived from TT and from pTT at <1 cm from the tumor border, in 40 patients with GBM. The expression of O6-methylguanine-DNA methyltransferase (MGMT), a suicidal DNA repair protein; Breast Cancer Resistance Protein (BCRP1), a drug efflux transporter, and A2B5 (c-series gangliosides) has been determined by immunohistochemistry. The percentage of MGMT positive cells was higher (p<0.0001, paired Student’s t test) in pTT (median: 53.5, range: 0.6-92.4) with respect to TT (median: 3.3, range: 0.0-70.7). The same trend was observed in BCRP1 expression (p<0.02; pTT, median: 27.6, range: 1.0-95.6; TT, median: 10.1, range: 0.2-72.1). No difference was found between pTT and TT in A2B5 expression (p=0.69, pTT, median: 29.8, range: 0.0-98.4; TT, median: 26.0, range: 0.0-96.8). Patients were then divided into two groups according to presence (group A) or absence (group B) of tumor cells in pTT. The trend previous observed in MGMT expression was maintained in both groups, while only in group A a statistically significant difference in BCRP1 expression was observed. Our results confirm that the tissue surrounding GBM is not a normal tissue, and that it represents a frontline of tumor invasion, particularly for the presence of some molecules involved in chemoresistance, which could explain the disease recurrence after the conventional treatment of GBM. Experiments about the expression of above mentioned molecules in CSC from pTT and TT are in progress. Supported by FIRB, ACCORDI DI PROGRAMMA 201

    Phospho-mTOR expression in human glioblastoma microglia-macrophage cells.

    Get PDF
    Abstract The glioblastoma (GBM) immune microenvironment is highly heterogeneous, and microglia may represent 30–70% of the entire tumor. However, the role of microglia and other specific immune populations is poorly characterized. Activation of mTOR signaling occurs in numerous human cancers and has roles in microglia-glioma cell interactions. We now show in human tumor specimens (42 patients), that 39% of tumor-associated microglial (TAM) cells express mTOR phosphorylated at Ser-2448; and similar mTOR activation is observed using a human microglia-glioma interaction paradigm. In addition, we confirm previous studies that microglia express urea and ARG1 (taken as M2 marker) in the presence of glioma cells, and this phenotype is down-regulated in the presence of a mTOR inhibitor. These results suggest that mTOR suppression in GBM patients might induce a reduction of the M2 phenotype expression in up to 40% of all TAMs. Since the M2 profile of microglial activation is believed to be associated with tumor progression, reductions in that phenotype may represent an additional anti-tumor mechanism of action of mTOR inhibitors, along with direct anti-proliferative activities

    Involvement of cancer stem cells in glioblastoma angiogenesis

    Get PDF
    It is widely accepted that glioblastoma (GBM) develops from cancer stem cells (CSCs), a subset of stem-like cells displaying high resistance to treatment. In fact, despite aggressive therapy, 90% of patients relapse within 2 cm from tumor edge. Our recent findings showed the existence of a CSC type, residing in GBM peritumor tissue (PCSCs), that bears distinct characteristics from CSCs of the tumor mass (GCSCs). It should be considered the possibility that, after surgical resection, PCSCs might represent a reservoir of cells able to recapitulate the tumor. In this setting, characterization of PCSCs appears to be crucial in order to identify novel effective therapeutic targets. Thus, our aim was to investigate GCSCs and PCSCs role in angiogenesis, a key event in both GBM and peritumor tissue, whose vasculature shows features similar to those found in the tumor mass. In particular, we analyzed, by immunocytochemistry (ICC), Western blotting or real-time PCR, the expression of molecules involved in hypoxia and angiogenesis, such as HIF1α, HIF2α, and VEGF along with its receptors (VEGFR1, VEGFR2). ICC has highlighted the presence and the specific localization of these molecules in both GCSCs and PCSCs. The two cell populations showed comparable levels of VEGF. The transcript of VEGFR1 was in general expressed at higher levels in GCSCs than in PCSCs, while VEGFR2 mRNA and protein did not show a unique trend of expression. The expression of VEGF and its receptors in both GCSCs and PCSCs suggests that, besides well-known paracrine loops, autocrine signalings are also involved in tumor angiogenesis. Moreover, the expression of angiogenesis markers in PCSCs suggests these cells to have a direct role in peritumor tissue new vessel formation. In this regard, PCSCs should be considered a promising therapeutic target to counteract the angiogenesis-supported tumor progression

    The Interference of Notch1 target Hes1 affects cell growth, differentiation and invasiveness of glioblastoma stem cells through modulation of multiple oncogenic targets

    Get PDF
    The invasive and lethal nature of Glioblastoma multiforme (GBM) necessitates the continuous identification of molecular targets and search of efficacious therapies to inhibit GBM growth. The GBM resistance to chemotherapy and radiation it is attributed to the existence of a rare fraction of cancer stem cells (CSC) that we have identified within the tumor core and in peritumor tissue of GBM. Since Notch1 pathway is a potential therapeutic target in brain cancer, earlier we highlighted that pharmacological inhibition of Notch1 signalling by γ-secretase inhibitor-X (GSI-X), reduced cell growth of some c-CSC than to their respective p-CSC, but produced negligible effects on cell cycle distribution, apoptosis and cell invasion. In the current study, we assessed the effects of Hes1-targeted shRNA, a Notch1 gene target, specifically on GBM CSC refractory to GSI-X. Depletion of Hes1 protein induces major changes in cell morphology, cell growth rate and in the invasive ability of shHes1-CSC in response to growth factor EGF. shHes1-CSC show a decrease of the stemness marker Nestin concurrently to a marked increase of neuronal marker MAP2 compared to pLKO.1-CSC. Those effects correlated with repression of EGFR protein and modulation of Stat3 phosphorylation at Y705 and S727 residues. In the last decade Stat3 has gained attention as therapeutic target in cancer but there is not yet any approved Stat3-based glioma therapy. Herein, we report that exposure to a Stat3/5 inhibitor, induced apoptosis either in shHes1-CSC or control cells. Taken together, Hes1 seems to be a favorable target but not sufficient itself to target GBM efficaciously, therefore a possible pharmacological intervention should provide for the use of anti-Stat3/5 drugs either alone or in combination regimen

    Glioblastoma stem cells express non-canonical proteins and exclusive mesenchymal-like or non-mesenchymal-like protein signatures

    Get PDF
    Glioblastoma (GBM) cancer stem cells (GSCs) contribute to GBM's origin, recurrence, and resistance to treatment. However, the understanding of how mRNA expression patterns of GBM subtypes are reflected at global proteome level in GSCs is limited. To characterize protein expression in GSCs, we performed in-depth proteogenomic analysis of patient-derived GSCs by RNA-sequencing and mass-spectrometry. We quantified > 10 000 proteins in two independent GSC panels and propose a GSC-associated proteomic signature characterizing two distinct phenotypic conditions; one defined by proteins upregulated in proneural and classical GSCs (GPC-like), and another by proteins upregulated in mesenchymal GSCs (GM-like). The GM-like protein set in GBM tissue was associated with necrosis, recurrence, and worse overall survival. Through proteogenomics, we discovered 252 non-canonical peptides in the GSCs, i.e., protein sequences that are variant or derive from genome regions previously considered non-protein-coding, including variants of the heterogeneous ribonucleoproteins implicated in RNA splicing. In summary, GSCs express two protein sets that have an inverse association with clinical outcomes in GBM. The discovery of non-canonical protein sequences questions existing gene models and pinpoints new protein targets for research in GBM
    corecore