Network


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

Hotspot


Dive into the research topics where Maria Laura Falchetti is active.

Publication


Featured researches published by Maria Laura Falchetti.


PLOS ONE | 2011

Mesenchymal Stromal Cells Primed with Paclitaxel Provide a New Approach for Cancer Therapy

Augusto Pessina; Arianna Bonomi; Valentina Coccè; Gloria Invernici; Stefania Elena Navone; Loredana Cavicchini; Francesca Sisto; Maura Ferrari; Lucia Viganò; Alberta Locatelli; Emilio Ciusani; Graziella Cappelletti; Daniele Cartelli; Caruso Arnaldo; Eugenio Parati; Giovanni Marfia; Roberto Pallini; Maria Laura Falchetti; Giulio Alessandri

Background Mesenchymal stromal cells may represent an ideal candidate to deliver anti-cancer drugs. In a previous study, we demonstrated that exposure of mouse bone marrow derived stromal cells to Doxorubicin led them to acquire anti-proliferative potential towards co-cultured haematopoietic stem cells (HSCs). We thus hypothesized whether freshly isolated human bone marrow Mesenchymal stem cells (hMSCs) and mature murine stromal cells (SR4987 line) primed in vitro with anti-cancer drugs and then localized near cancer cells, could inhibit proliferation. Methods and Principal Findings Paclitaxel (PTX) was used to prime culture of hMSCs and SR4987. Incorporation of PTX into hMSCs was studied by using FICT-labelled-PTX and analyzed by FACS and confocal microscopy. Release of PTX in culture medium by PTX primed hMSCs (hMSCsPTX) was investigated by HPLC. Culture of Endothelial cells (ECs) and aorta ring assay were used to test the anti-angiogenic activity of hMSCsPTX and PTX primed SR4987(SR4987PTX), while anti-tumor activity was tested in vitro on the proliferation of different tumor cell lines and in vivo by co-transplanting hMSCsPTX and SR4987PTX with cancer cells in mice. Nevertheless, despite a loss of cells due to chemo-induced apoptosis, both hMSCs and SR4987 were able to rapidly incorporate PTX and could slowly release PTX in the culture medium in a time dependent manner. PTX primed cells acquired a potent anti-tumor and anti-angiogenic activity in vitro that was dose dependent, and demonstrable by using their conditioned medium or by co-culture assay. Finally, hMSCsPTX and SR4987PTX co-injected with human cancer cells (DU145 and U87MG) and mouse melanoma cells (B16) in immunodeficient and in syngenic mice significantly delayed tumor takes and reduced tumor growth. Conclusions These data demonstrate, for the first time, that without any genetic manipulation, mesenchymal stromal cells can uptake and subsequently slowly release PTX. This may lead to potential new tools to increase efficacy of cancer therapy.


Neurosurgery | 2005

Homologous transplantation of neural stem cells to the injured spinal cord of mice.

Roberto Pallini; Lucia Ricci Vitiani; Alessandra Bez; Patrizia Casalbore; Francesco Facchiano; Valeria Di Giorgi Gerevini; Maria Laura Falchetti; Eduardo Fernandez; Giulio Maira; Cesare Peschle; Eugenio Parati

OBJECTIVE:Murine neural stem cells (NSCs) were homografted onto the injured spinal cord (SC) to assess their potential to improve motor behavior, to differentiate as neurons, and to establish synapse-like contacts with the descending axonal paths of the host. In addition, we investigated whether transduced NSCs over-expressing vascular endothelial growth factor might exert any angiogenetic effect in the injured SC. METHODS:NSCs derived from mouse embryos were transduced to express either green fluorescent protein or vascular endothelial growth factor. The cells were engrafted in mice where an extended dorsal funiculotomy had been performed at the T8–T9 level. At intervals from 4 to 12 weeks after grafting, motor behavior was assessed using an open field locomotor scale and footprint analysis. At the same time points, the SC was studied by conventional histology, immunohistochemistry, and fluorescence microscopy. The interactions between the grafted NSCs and descending axonal paths were investigated using anterogradely transported fluorescent axonal tracers. RESULTS:By the 12–week time point, mice engrafted with NSCs significantly improved both their locomotor score on open field test and their base of support on footprint analysis. Histological studies showed that green fluorescent protein-positive NSCs survived as long as 12 weeks after grafting, migrated from the grafting site with a tropism toward the lesion, and either remained undifferentiated or differentiated into the astrocytic phenotype without neuronal or oligodendrocytic differentiation. Interestingly, the NSC-derived astrocytes expressed vimentin, suggesting that these cells differentiated as immature astrocytes. The tips of severed descending axonal paths came adjacent to grafted NSCs without forming synapse-like structures. When genetically engineered to over-express vascular endothelial growth factor, the grafted NSCs significantly increased vessel density in the injured area. CONCLUSION:In the traumatically injured mice SC, NSC grafting improves motor recovery. Although differentiation of engrafted NSCs is restricted exclusively toward the astrocytic phenotype, the NSC-derived astrocytes show features that are typical of the early phase after SC injury when the glial scar is still permissive to regenerating axons. The immature phenotype of the NSC-derived astrocytes suggests that these cells might support neurite outgrowth by the host neurons. Thus, modifying the glial scar with NSCs might enhance axonal regeneration in the injured area. The use of genetically engineered NSCs that express trophic factors appears to be an attractive tool in SC transplantation research.


International Journal of Cancer | 2000

In situ detection of telomerase catalytic subunit mRNA in glioblastoma multiforme

Maria Laura Falchetti; Roberto Pallini; Ettore D'Ambrosio; Francesco Pierconti; Maurizio Martini; Graziella Cimino-Reale; Roberto Verna; Giulio Maira; Luigi Maria Larocca

Activation of telomerase may allow unlimited cell proliferation and immortalization. One of the telomerase protein subunits has a reverse transcriptase (hTERT) activity that is essential for telomerase function and regulation. In human gliomas, telomerase is frequently associated with malignant tumor progression. In our study, we investigated the expression of hTERT at the cellular level in 34 primary de novo glioblastoma multiforme (GBM) by in situ hybridization ( ISH ). The expression of hTERT in tumor tissue was also assessed by RT‐PCR. In addition, telomerase activity measured by telomeric repeat amplification protocol (TRAP) and telomere length polymorphism assayed by telomere restriction fragment (TRF) Southern blot were investigated. We found that all GBM, including those with negative TRAP reaction, contained abundant amounts of cytoplasmic hTERT mRNA. Interestingly, the ISH analysis revealed that the hTERT mRNA was homogeneously expressed by the whole tumor cell population in about 60% of the GBM. In the remaining cases, hTERT was absent in subsets of tumor cells. TRF analysis, which shows that both TRAP‐positive and TRAP‐negative de novo GBM have elongated telomeres, further supports that telomerase activity is present in all de novo GBM. Correlations with tumor size and extent of necrosis suggest that hTERT reactivation is an early event in GBM development and that telomerase activity may be lost in subpopulations of neoplastic cells during tumor progression. Finally, ISH analysis of hTERT mRNA seems to provide a prognostic parameter for primary de novo GBM. Int. J. Cancer 88:895–901, 2000.


Neurological Research | 2006

Influence of local environment on the differentiation of neural stem cells engrafted onto the injured spinal cord.

Lucia Ricci-Vitiani; Patrizia Casalbore; Giovanna Petrucci; Liverana Lauretti; Nicola Montano; Luigi Maria Larocca; Maria Laura Falchetti; Dario Lombardi; Valeria Di Giorgio Gerevini; Carlo Cenciarelli; Quintino Giorgio D'Alessandris; Eduardo Fernandez; Ruggero De Maria; Giulio Maira; Cesare Peschle; Eugenio Parati; Roberto Pallini

Abstract Objectives: In vitro, neural stem cells (NSCs) proliferate as undifferentiated spheroids and differentiate into neurons, astrocytes and oligodendrocytes. These features make NSCs suitable for spinal cord (SC) reconstruction. However, in vivo experiments have demonstrated that in the injured SC transplanted NSCs either remain undifferentiated or differentiate into the astrocytic phenotype. The microenvironment of the injured SC is believed to play a crucial role in driving the differentiation of the engrafted NSCs. Here, we tested the hypothesis that inflammatory cytokines (ICs) may be involved in the restricted differentiation of NSCs after grafting onto the injured SC. Methods: As the first step, we used immunohistochemistry to analyse the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and interferon (IFN)-γ in the normal SC of mice and following traumatic injury. Then, we investigated whether a combination of TNF-α, IL-1β and IFN-γ may affect the phenotype of murine NSCs in vitro. Results: We found that TNF-α, IL–1β and IFN-γ, which are absent in the normal SC, are all expressed in the injured SC and the expression of these cytokines follows a timely tuned fashion with IFN-γ being detectable as long as 4 weeks after injury. In culture, exposure of proliferating NSCs to a combination of TNF-α, IL–1β and IFN-γ was per se sufficient to induce the astrocytic differentiation of these cells even in the absence of serum. Conclusions: In the traumatically injured SC, differentiation of engrafted NSCs is restricted towards the astrocytic lineage because of the inflammatory environment. ICs are likely to play a major role in differentiation of NSCs in the in vivo conditions.


International Journal of Cancer | 2008

Inhibition of telomerase in the endothelial cells disrupts tumor angiogenesis in glioblastoma xenografts

Maria Laura Falchetti; Maria Patrizia Mongiardi; Paolo Fiorenzo; Giovanna Petrucci; Francesco Pierconti; Igea D'Agnano; Giorgio D'Alessandris; Giulio Alessandri; Maurizio Gelati; Lucia Ricci-Vitiani; Giulio Maira; Luigi Maria Larocca; Andrea Levi; Roberto Pallini

Tumor angiogenesis is a complex process that involves a series of interactions between tumor cells and endothelial cells (ECs). In vitro, glioblastoma multiforme (GBM) cells are known to induce an increase in proliferation, migration and tube formation by the ECs. We have previously shown that in human GBM specimens the proliferating ECs of the tumor vasculature express the catalytic component of telomerase, hTERT, and that telomerase can be upregulated in human ECs by exposing these cells to GBM in vitro. Here, we developed a controlled in vivo assay of tumor angiogenesis in which primary human umbilical vascular endothelial cells (HUVECs) were subcutaneously grafted with or without human GBM cells in immunocompromised mice as Matrigel implants. We found that primary HUVECs did not survive in Matrigel implants, and that telomerase upregulation had little effect on HUVEC survival. In the presence of GBM cells, however, the grafted HUVECs not only survived in Matrigel implants but developed tubule structures that integrated with murine microvessels. Telomerase upregulation in HUVECs enhanced such effect. More importantly, inhibition of telomerase in HUVECs completely abolished tubule formation and greatly reduced survival of these cells in the tumor xenografts. Our data demonstrate that telomerase upregulation by the ECs is a key requisite for GBM tumor angiogenesis.


International Journal of Cancer | 2006

Telomerase inhibition by stable RNA interference impairs tumor growth and angiogenesis in glioblastoma xenografts

Roberto Pallini; Antonio Sorrentino; Francesco Pierconti; Nicola Maggiano; Riccardo Faggi; Nicola Montano; Giulio Maira; Luigi Maria Larocca; Andrea Levi; Maria Laura Falchetti

Telomerase is highly expressed in advanced stages of most cancers where it allows the clonal expansion of transformed cells by counteracting telomere erosion. Telomerase may also contribute to tumor progression through still undefined cell growth‐promoting functions. Here, we inhibited telomerase activity in 2 human glioblastoma (GBM) cell lines, TB10 and U87MG, by targeting the catalytic subunit, hTERT, via stable RNA interference (RNAi). Although the reduction in telomerase activity had no effect on GBM cell growth in vitro, the development of tumors in subcutaneously and intracranially grafted nude mice was significantly inhibited by antitelomerase RNAi. The in vivo effect was observed within a relatively small number of population doublings, suggesting that telomerase inhibition may hinder cancer cell growth in vivo prior to a substantial shortening of telomere length. Tumor xenografts that arose from telomerase‐inhibited GBM cells also showed a less‐malignant phenotype due both to the absence of massive necrosis and to reduced angiogenesis.


Oncogene | 1999

Induction of telomerase activity in v-myc-transformed avian cells

Maria Laura Falchetti; Germana Falcone; Ettore D'Ambrosio; Roberto Verna; Stefano Alemà; Andrea Levi

Telomerase activity is detectable in the majority of tumors or immortalized cell lines, but is repressed in most normal human somatic cells. It is generally assumed that reactivation of telomerase prevents the erosion of chromosome ends which occurs in cycling cells and, hence, hinders cellular replicative senescence. Here, we show that the expression of v-Myc oncoprotein by retroviral infection of telomerase-negative embryonal quail myoblasts and chicken neuroretina cells is sufficient for reactivating telomerase activity, earlier than telomere shortening could occur. Furthermore, the use of a conditional v-Myc-estrogen receptor protein (v-MycER) causes estrogen-dependent expression of detectable levels of telomerase activity in recently infected chick embryo fibroblasts and neuroretina cells. We conclude that the high levels of telomerase activity in v-Myc-expressing avian cells are not the mere consequence of transformation or of a differentiative block, since v-Src tyrosine kinase, which prevents terminal differentiation and promotes cell transformation, fails to induce telomerase activity.


British Journal of Haematology | 2013

Mesenchymal stromal cells primed with Paclitaxel attract and kill leukaemia cells, inhibit angiogenesis and improve survival of leukaemia‐bearing mice

Augusto Pessina; Valentina Coccè; Luisa Pascucci; Arianna Bonomi; Loredana Cavicchini; Francesca Sisto; Maura Ferrari; Emilio Ciusani; Antonio Crovace; Maria Laura Falchetti; Sonia Zicari; Arnaldo Caruso; Stefania Elena Navone; Giovanni Marfia; Anna Benetti; P. Ceccarelli; Eugenio Parati; Giulio Alessandri

Current leukaemia therapy focuses on increasing chemotherapy efficacy. Mesenchymal stromal cells (MSCs) have been proposed for carrying and delivery drugs to improve killing of cancer cells. We have shown that MSCs loaded with Paclitaxel (PTX) acquire a potent anti‐tumour activity. We investigated the effect of human MSCs (hMSCs) and mouse SR4987 loaded with PTX (hMSCsPTX and SR4987PTX) on MOLT‐4 and L1210, two leukaemia cell (LCs) lines of human and mouse origin, respectively. SR4987PTX and hMSCsPTX showed strong anti‐LC activity. hMSCsPTX, co‐injected with MOLT‐4 cells or intra‐tumour injected into established subcutaneous MOLT‐4 nodules, strongly inhibited growth and angiogenesis. In BDF1‐mice‐bearing L1210, the intraperitoneal administration of SR4987PTX doubled mouse survival time. In vitro, both hMSCs and hMSCsPTX released chemotactic factors, bound and formed rosettes with LCs. In ultrastructural analysis of rosettes, hMSCsPTX showed no morphological alterations while the attached LCs were apoptotic and necrotic. hMSCs and hMSCsPTX released molecules that reduced LC adhesion to microvascular endothelium (hMECs) and down‐modulated ICAM1 and VCAM1 on hMECs. Priming hMSCs with PTX is a simple procedure that does not require any genetic cell manipulation. Once the effectiveness of hMSCsPTX on established cancers in mice is proven, this procedure could be proposed for leukaemia therapy in humans.


American Journal of Clinical Pathology | 2012

Prognostic Relevance of c-Myc and BMI1 Expression in Patients With Glioblastoma

Tonia Cenci; Maurizio Martini; Nicola Montano; Quintino Giorgio D’Alessandris; Maria Laura Falchetti; Daniela Annibali; Mauro Savino; Federico Bianchi; Francesco Pierconti; Sergio Nasi; Roberto Pallini; Luigi Maria Larocca

Although the c-Myc oncogene is frequently deregulated in human cancer, its involvement in the pathogenesis of glioblastoma is not clear. We conducted immunohistochemical analysis of the expression of c-Myc, polycomb ring finger oncogene (BMI1), and acetylation of the lysine 9 (H3K9Ac) of histone 3 in 48 patients with glioblastoma who underwent surgery followed by radiotherapy and temozolomide treatment. The expression of c-Myc, BMI1, and H3K9ac was correlated with clinical characteristics and outcome. We found that overexpression of c-Myc was significantly associated with that of BMI1 (P = .009), and that patients who harbored glioblastomas overexpressing c-Myc and BMI1 showed significantly longer overall survival (P < .0001 and P = .0009, respectively). Our results provide the first evidence of the prognostic value of c-Myc and associated genes in patients with glioblastoma. The favorable effect of c-Myc and BMI1 expression on survival is likely mediated by the sensitization of cancer cells to radiotherapy and temozolomide through the activation of apoptotic pathways.


Stem Cell Research & Therapy | 2014

Immortalization of human adipose-derived stromal cells: production of cell lines with high growth rate, mesenchymal marker expression and capability to secrete high levels of angiogenic factors

Luigi Balducci; Antonella Blasi; Marilisa Saldarelli; Antonio Soleti; Augusto Pessina; Arianna Bonomi; Valentina Coccè; Marta Dossena; Valentina Tosetti; Valentina Ceserani; Stefania Elena Navone; Maria Laura Falchetti; Eugenio Parati; Giulio Alessandri

IntroductionHuman adipose-derived stromal cells (hASCs), due to their relative feasibility of isolation and ability to secrete large amounts of angiogenic factors, are being evaluated for regenerative medicine. However, their limited culture life span may represent an obstacle for both preclinical investigation and therapeutic use. To overcome this problem, hASCs immortalization was performed in order to obtain cells with in vitro prolonged life span but still maintain their mesenchymal marker expression and ability to secrete angiogenic factors.MethodshASCs were transduced with the human telomerase reverse transcriptase (hTERT) gene alone or in combination with either SV-40 or HPV E6/E7 genes. Mesenchymal marker expression on immortalized hASCs lines was confirmed by flow cytometry (FC), differentiation potential was evaluated by immunocytochemistry and ELISA kits were used for evaluation of angiogenic factors. Green fluorescent protein (GFP) gene transduction was used to obtain fluorescent cells.ResultsWe found that hTERT alone failed to immortalize hASCs (hASCs-T), while hTERT/SV40 (hASCs-TS) or hTERT/HPV E6/E7 (hASCs-TE) co-transductions successfully immortalized cells. Both hASCs-TS and hASCs-TE were cultured for up to one year with a population doubling level (PDL) up to 100. Comparative studies between parental not transduced (hASCs-M) and immortalized cell lines showed that both hASCs-TS and hASCs-TE maintained a mesenchymal phenotypic profile, whereas differentiation properties were reduced particularly in hASCs-TS. Interestingly, hASCs-TS and hASCs-TE showed a capability to secrete significant amount of HGF and VEGF. Furthermore, hASCs-TS and hASCs-TE did not show tumorigenic properties in vitr o although some chromosomal aberrations were detected. Finally, hASCs-TS and hASCs-TE lines were stably fluorescent upon transduction with the GFP gene.ConclusionsHere we demonstrated, for the first time, that hASCs, upon immortalization, maintain a strong capacity to secrete potent angiogenic molecules. By combining hASCs immortalization and their paracrine characteristics, we have developed a “hybridoma-like model” of hASCs that could have potential applications for discovering and producing molecules to use in regenerative medicine (process scale-up).In addition, due to the versatility of these fluorescent-immortalized cells, they could be employed in in vivo cell-tracking experiments, expanding their potential use in laboratory practice.

Collaboration


Dive into the Maria Laura Falchetti's collaboration.

Top Co-Authors

Avatar

Roberto Pallini

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Luigi Maria Larocca

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Giulio Maira

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Andrea Levi

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Francesco Pierconti

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eugenio Parati

Carlo Besta Neurological Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge