Umberto Galderisi

Professor of Molecular Biology

Name Umberto
Surname Galderisi
Institution Università degli Studi della Campania Luigi Vanvitelli
Telephone +39 081 566 75 85
E-Mail umberto.galderisi@unicampania.it
Address Dept. of Experimental Medicine, Via Luigi De Crecchio 7 – 80138 Napoli, Italy
Umberto Galderisi

Member PUBLICATIONS

  • Reduced expression of MECP2 affects cell commitment and maintenance in neurons by triggering senescence: new perspective for Rett syndrome.

    Publication Date: 01/04/2012 on Molecular biology of the cell
    by Squillaro T, Alessio N, Cipollaro M, Melone MA, Hayek G, Renieri A, Giordano A, Galderisi U
    DOI: 10.1091/mbc.E11-09-0784

    MECP2 protein binds preferentially to methylated CpGs and regulates gene expression by causing changes in chromatin structure. The mechanism by which impaired MECP2 activity can induce pathological abnormalities in the nervous system of patients with Rett syndrome (RTT) is not clearly understood. To gain further insight into the role of MECP2 in human neurogenesis, we compared the neural differentiation process in mesenchymal stem cells (MSCs) obtained from a RTT patient and from healthy donors. We further analyzed neural differentiation in a human neuroblastoma cell line carrying a partially silenced MECP2 gene. Senescence and reduced expression of neural markers were observed in proliferating and differentiating MSCs from the RTT patient, which suggests that impaired activity of MECP2 protein may impair neural differentiation, as observed in RTT patients. Next, we used an inducible expression system to silence MECP2 in neuroblastoma cells before and after the induction of neural differentiation via retinoic acid treatment. This approach was used to test whether MECP2 inactivation affected the cell fate of neural progenitors and/or neuronal differentiation and maintenance. Overall, our data suggest that neural cell fate and neuronal maintenance may be perturbed by senescence triggered by impaired MECP2 activity either before or after neural differentiation.

  • Stem cell therapy for arterial restenosis: potential parameters contributing to the success of bone marrow-derived mesenchymal stromal cells.

    Publication Date: 01/02/2012 on Cardiovascular drugs and therapy
    by Forte A, Rinaldi B, Sodano L, Berrino L, Rossi F, Finicelli M, Grossi M, Cobellis G, Botti C, De Feo M, Santè P, Galderisi U, Cipollaro M
    DOI: 10.1007/s10557-011-6359-8

    Restenosis is a complex and heterogeneous pathophysiological phenomenon occurring in patients submitted to revascularization procedures. Previous studies proved the antirestenotic properties of injected allogenic mesenchymal stromal cells (MSCs) in an experimental model of rat carotid (re)stenosis induced through arteriotomy. In this study we describe some of the effects subsequent to MSC treatment of rats submitted to carotid arteriotomy and possibly responsible for their antirestenotic effect.

  • Chromatin modification and senescence.

    Publication Date: 01/01/2012 on Current pharmaceutical design
    by Di Bernardo G, Cipollaro M, Galderisi U

    Cells are the fundamental structure composing our bodies and hence cellular decline (called senescence) contributes to ageing. Endogenous and exogenous stresses may induce cellular senescence. Stressors are mainly macromolecule damage events, which include: shortening of chromosome telomeres; non-telomeric DNA damage; excessive mitogenic signals, which may cause DNA damage; and non-genotoxic stress, such as perturbations to chromatin organization. For many years the analysis of chromatin perturbation as a leading event in triggering senescence has been overlooked. Now, it is well recognized that chromatin DNA packaging is not immune to the ravages of time. All eukaryotes experience changes in chromatin organization and gene-expression patterns as they age. This can be due to perturbation in the function of chromatin modifiers. In this review we will discuss the role in the senescence process of the different types of chromatin modifiers, such as the ATP-dependent chromatin remodelling complexes, the enzymes that covalently modify histone tails and proteins involved in DNA methylation.

  • Morphological and molecular characterization of healthy human ascending aorta.

    Publication Date: 01/01/2012 on Histology and histopathology
    by Forte A, Della Corte A, Grossi M, Finicelli M, Bancone C, Provenzano R, Pepino P, Nappi GA, De Feo M, Galderisi U, Cotrufo M, Cipollaro M
    DOI: 10.14670/HH-27.103

    Knowledge of the characteristics of the normal human aorta has been constrained by lack of data on fresh aortic tissue, especially from healthy individuals. In this study, the gene expression and morphological characteristics of the thoracic ascending aorta (AA) of healthy organ donors have been evaluated, with the aim of providing reference data for the analysis of pathological AAs. We analysed by RT-PCR the differential expression of mRNAs coding for myocardin, smoothelin, alpha-smooth muscle actin (alpha-SMA) and the ED-A isoform of fibronectin (ED-A FN) in AA specimens from donors, integrating the results with immunohistochemical analysis of the same targets. Morphological and morphometric characteristics of the AAs were also evaluated. In order to account for possible regional variations in wall structure, the convexity of the aortic profile was compared to the concavity. No differences in gene expression occurred for any of the target genes between the concavity and the convexity of AAs. Immunohistochemistry revealed a different distribution of total FN and of its ED-A isoform in the media and in the intima. Smoothelin is expressed by the majority of cells in the media, with some positive cells also in the intima. Alpha-SMA is expressed in all the tunicae. Immunohistochemistry also revealed in the convexity of 50% of AAs the presence of discrete areas in the subadventital media with altered structure and cell morphology and with altered gene expression, resulting positive for ED-A FN and alpha-SMA, but not for smoothelin, indicating the occurrence of early lesions also in macroscopically healthy AAs.

  • Impact of histone deacetylase inhibitors SAHA and MS-275 on DNA repair pathways in human mesenchymal stem cells.

    Publication Date: 01/11/2010 on Journal of cellular physiology
    by Di Bernardo G, Alessio N, Dell'Aversana C, Casale F, Teti D, Cipollaro M, Altucci L, Galderisi U
    DOI: 10.1002/jcp.22236

    Histone deacetylase inhibitors (HDACis) have received considerable attention for their anti-tumoral properties. We report here the effects of two HDACis, SAHA and MS-275, on the biology of mesenchymal stem cells (MSCs). It is well known that HDACis trigger both DNA damage responses and actual DNA damage in cancer cells. On this premise, we evaluated HDACis influence on DNA damage pathways in MSCs. We analyzed a panel of genes involved in the regulation of base and nucleotide excision repair, mismatch repair, and double strand break repair. That a majority of the analyzed genes displayed significant expression changes upon incubation with SAHA or MS-275 suggested that regulation of their expression is greatly affected by HDACis. The complex expression pattern, with some genes up-regulated and other under-expressed, did not allow to foresee whether these changes allow cells cope with stressful DNA damaging stimuli. Furthermore, we evaluated the biological outcome following treatment of MSCs with DNA damaging agents (H(2)O(2) and UV) in presence of HDACis. In these settings, MSCs treated with H(2)O(2) or UV radiation underwent apoptosis and/or senescence, and pre-incubation with HDACi exacerbated cell death phenomena. Accordingly, the number of cells harboring 8-oxo-7,8-dihydroguanine (8oxodG), a hallmark of DNA oxidative damage, was significantly higher in samples incubated with HDACis compared to controls. In summary, our findings suggest that SAHA and MS-275, even at low effective doses, can alter the biology of MSCs, diminishing their ability to survive the effects of DNA-damaging agents.

  • Human mesenchymal stem cells as novel neuropathic pain tool.

    Publication Date: 23/10/2010 on Journal of stem cells & regenerative medicine
    by Siniscalco D, Giordano C, Galderisi U, Luongo L, Alessio N, Di Bernardo G, de Novellis V, Rossi F, Maione S
  • The BRG1 ATPase of chromatin remodeling complexes is involved in modulation of mesenchymal stem cell senescence through RB-P53 pathways.

    Publication Date: 07/10/2010 on Oncogene
    by Alessio N, Squillaro T, Cipollaro M, Bagella L, Giordano A, Galderisi U
    DOI: 10.1038/onc.2010.285

    We focused our attention on brahma-related gene 1 (BRG1), the ATPase subunit of the SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex, and analyzed its role in mesenchymal stem cell (MSC) biology. We hypothesized that deviation from the correct concentration of these proteins, which act at the highest level of gene regulation, may be deleterious for cells. We wanted to know what would happen if a cell had to cope with altered regulation of gene expression, either by upregulation or downregulation of BRG1. We assumed that cells would try to restore homeostasis or, alternatively, that the event could trigger senescence/apoptosis phenomena. To this end, in MSCs, we silenced BRG1gene. Knockdown of BRG1 expression induced a significant increase in senescent cells and decrease in apoptotic cells. It is interesting that BRG1 downregulation also induced an increase in heterochromatin. At the molecular level, these phenomena were associated with activation of retinoblastoma-like protein 2 (RB2)/P130- and P53-related pathways. Senescence was accompanied by reduced expression of some stemness-related genes. This is consistent with our previous research, which showed that BRG1 upregulation by ectopic expression also induced senescence processes. Together, these data suggest that BRG1 belongs to a class of genes whose expression is tightly regulated; hence, subtle alterations in BRG1 activity seem to negatively affect mechanisms regulating chromatin status and, in turn, impair cellular physiology.

  • Spontaneous resolution of eosinophilic granuloma in a patient with a psychotic disorder.

    Publication Date: 01/09/2010 on The neuroradiology journal
    by Conforti R, Porto A, Cirillo M, Sgambato A, Galderisi S, Cirillo S
    DOI: 10.1177/197140091002300412

    A 16-year-old female who manifested psychotic symptoms underwent CT and MRI for the evaluation of an incidentally discovered asymptomatic palpable mass of the right occipital region of the skull. The correlation between clinical and radiological data and biopsy data led to the diagnosis of eosinophilic granuloma. The radiological finding is discussed and reviewed in relation to clinical aspects and literature data.

  • Controlled delivery of the heparan sulfate/FGF-2 complex by a polyelectrolyte scaffold promotes maximal hMSC proliferation and differentiation.

    Publication Date: 01/07/2010 on Journal of cellular biochemistry
    by Calarco A, Petillo O, Bosetti M, Torpedine A, Cannas M, Perrone L, Galderisi U, Melone MA, Peluso G
    DOI: 10.1002/jcb.22602

    Growth factors and other regulatory molecules are required to direct differentiation of bone marrow-derived human mesenchymal stem cells (hMSC) along specific lineages. However, the therapeutic use of growth factors is limited by their susceptibility to degradation, and the need to maintain prolonged local release of growth factor at levels sufficient to stimulate hMSC. The aim of this study was to investigate whether a device containing heparan sulfate (HS), which is a co-factor in growth factor-mediated cell proliferation and differentiation, could potentiate and prolong the delivery of fibroblast growth factor-2 (FGF-2) and thus enhance hMSC stimulation. To this aim, we synthesized cationic polyelectrolyte polymers covalently and non-covalently anchored to HS and evaluated their effect on hMSC proliferation. Polymers non-covalently bound to HS resulted in the release of an HS/FGF-2 complex rather than FGF-2 alone. The release of this complex significantly restored hMSC proliferation, which was abolished in serum-free medium and only partially restored by the release of FGF-2 alone as occurred with polymer covalently bound to HS. We also demonstrate that exposure to HS/FGF-2 during early growth but not during post-confluence is essential for hMSC differentiation down the fibroblast lineage, which suggests that both factors are required to establish the correct stem cell commitment that is necessary to support subsequent differentiation. In conclusion, the delivery platform described here is a step towards the development of a new class of biomaterial that enables the prolonged, non-covalent binding and controlled delivery of growth factors and cofactors without altering their potency.

  • Partial silencing of methyl cytosine protein binding 2 (MECP2) in mesenchymal stem cells induces senescence with an increase in damaged DNA.

    Publication Date: 01/05/2010 on FASEB journal : official publication of the Federation of American Societies for Experimental Biology
    by Squillaro T, Alessio N, Cipollaro M, Renieri A, Giordano A, Galderisi U
    DOI: 10.1096/fj.09-143057

    DNA methylation is an epigenetic modification that occurs almost exclusively on CpG dinucleotides. MECP2 is a member of a family of proteins that preferentially bind to methylated CpGs. We analyzed the contribution of MECP2 to the physiology of mesenchymal stem cells (MSCs). Partial silencing of MECP2 in human MSCs induced a significant reduction of S-phase cells, along with an increase in G(1) cells. These changes were accompanied by a reduction of apoptosis, the triggering of senescence, a decrease in telomerase activity, and the down-regulation of genes involved in maintaining stem cell properties. Senescence appeared to rely on impairment of DNA damage repair and seemed to occur through RB- and P53-related pathways. The effects of MECP2 silencing could be related to the modification of the DNA methylation status. Our results indicate that the silencing of MECP2 induces an increase in methylated cytosines in the genome. Nevertheless, MECP2 partial silencing did not change the methylation of promoters, whose expression is affected by MECP2 down-regulation.