Network


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

Hotspot


Dive into the research topics where Giulia Grisendi is active.

Publication


Featured researches published by Giulia Grisendi.


Cancer Research | 2010

Adipose-derived mesenchymal stem cells as stable source of tumor necrosis factor-related apoptosis-inducing ligand delivery for cancer therapy.

Giulia Grisendi; Rita Bussolari; Luigi Cafarelli; István Peták; Valeria Rasini; Elena Veronesi; Giorgio De Santis; Carlotta Spano; Mara Tagliazzucchi; Helga Barti-Juhasz; Laura Scarabelli; Franco Bambi; Antonio Frassoldati; Giulio Rossi; Christian Casali; Uliano Morandi; Edwin M. Horwitz; Paolo Paolucci; Pierfranco Conte; Massimo Dominici

Adipose-derived mesenchymal stromal/stem cells (AD-MSC) may offer efficient tools for cell-based gene therapy approaches. In this study, we evaluated whether AD-MSC could deliver proapoptotic molecules for cancer treatment. Human AD-MSCs were isolated and transduced with a retroviral vector encoding full-length human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a proapoptotic ligand that induces apoptosis in a variety of human cancers but not normal tissues. Although several studies have documented the antitumor activity of recombinant human TRAIL, its use in vivo is limited by a short half-life in plasma due to a rapid clearance by the kidney. We found that these limitations can be overcome using stably transduced AD-MSC, which could serve as a constant source of TRAIL production. AD-MSC armed with TRAIL targeted a variety of tumor cell lines in vitro, including human cervical carcinoma, pancreatic cancer, colon cancer, and, in combination with bortezomib, TRAIL-resistant breast cancer cells. Killing activity was associated with activation of caspase-8 as expected. When injected i.v. or s.c. into mice, AD-MSC armed with TRAIL localized into tumors and mediated apoptosis without significant apparent toxicities to normal tissues. Collectively, our results provide preclinical support for a model of TRAIL-based cancer therapy relying on the use of adipose-derived mesenchymal progenitors as cellular vectors.


Cancer Research | 2013

Inhibiting Interactions of Lysine Demethylase LSD1 with Snail/Slug Blocks Cancer Cell Invasion

Giovanna Ferrari-Amorotti; Valentina Fragliasso; Roza Esteki; Zelia Prudente; Angela Rachele Soliera; Sara Cattelani; Gloria Manzotti; Giulia Grisendi; Massimo Dominici; Marco Pieraccioli; Giuseppe Raschellà; Claudia Chiodoni; Mario P. Colombo; Bruno Calabretta

The process of epithelial-mesenchymal transition (EMT) which is required for cancer cell invasion is regulated by a family of E-box-binding transcription repressors, which include Snail (SNAIL1) and Slug (SNAI2). Snail appears to repress the expression of the EMT marker E-cadherin by epigenetic mechanisms dependent on the interaction of its N-terminal SNAG domain with chromatin-modifying proteins including lysine-specific demethylase 1 (LSD1/KDM1A). We assessed whether blocking Snail/Slug-LSD1 interaction by treatment with Parnate, an enzymatic inhibitor of LSD1, or TAT-SNAG, a cell-permeable peptide corresponding to the SNAG domain of Slug, suppresses the motility and invasiveness of cancer cells of different origin and genetic background. We show here that either treatment blocked Slug-dependent repression of the E-cadherin promoter and inhibited the motility and invasion of tumor cell lines without any effect on their proliferation. These effects correlated with induction of epithelial and repression of mesenchymal markers and were phenocopied by LSD1 or Slug downregulation. Parnate treatment also inhibited bone marrow homing/engraftment of Slug-expressing K562 cells. Together, these studies support the concept that targeting Snail/Slug-dependent transcription repression complexes may lead to the development of novel drugs selectively inhibiting the invasive potential of cancer cells.


BMC Medicine | 2015

Mesenchymal stem/stromal cells as a delivery platform in cell and gene therapies

Naomi D’souza; Filippo Rossignoli; Giulia Golinelli; Giulia Grisendi; Carlotta Spano; Olivia Candini; Satoru Osturu; Fabio Catani; Paolo Paolucci; Edwin M. Horwitz; Massimo Dominici

Regenerative medicine relying on cell and gene therapies is one of the most promising approaches to repair tissues. Multipotent mesenchymal stem/stromal cells (MSC), a population of progenitors committing into mesoderm lineages, are progressively demonstrating therapeutic capabilities far beyond their differentiation capacities. The mechanisms by which MSC exert these actions include the release of biomolecules with anti-inflammatory, immunomodulating, anti-fibrogenic, and trophic functions. While we expect the spectra of these molecules with a therapeutic profile to progressively expand, several human pathological conditions have begun to benefit from these biomolecule-delivering properties. In addition, MSC have also been proposed to vehicle genes capable of further empowering these functions. This review deals with the therapeutic properties of MSC, focusing on their ability to secrete naturally produced or gene-induced factors that can be used in the treatment of kidney, lung, heart, liver, pancreas, nervous system, and skeletal diseases. We specifically focus on the different modalities by which MSC can exert these functions. We aim to provide an updated understanding of these paracrine mechanisms as a prerequisite to broadening the therapeutic potential and clinical impact of MSC.


Cytotherapy | 2010

GMP-manufactured density gradient media for optimized mesenchymal stromal/stem cell isolation and expansion

Giulia Grisendi; Cecilia Annerén; Luigi Cafarelli; Rita Sternieri; Elena Veronesi; Gian Luca Cervo; Stefano Luminari; Michela Maur; Antonio Frassoldati; Giovanni Palazzi; Satoru Otsuru; Franco Bambi; Paolo Paolucci; Conte Pierfranco; Edwin M. Horwitz; Massimo Dominici

BACKGROUND AIMS Bone marrow (BM) mesenchymal stromal/stem cells (MSC) are therapeutic tools in regenerative medicine and oncology. MSC isolation is often performed starting from a separation step based on research-grade 1.077 g/mL density gradient media (DGM). However, MSC clinical application should require the introduction of good manufacturing practice (GMP) reagents. We took advantage of two novel GMP DGM with densities of 1.077 and 1.073 g/mL (Ficoll-Paque PREMIUM and Ficoll-Paque PREMIUM 1.073, respectively) to test whether these reagents could isolate MSC efficiently while simultaneously comparing their performance. METHODS BM samples were processed using either 1.077 or 1.073 g/mL GMP DGM. BM mononucleated cell (MNC) fractions were analyzed for viability, immunophenotype, clonogenic potential, ex vivo expansion and differentiation potential. RESULTS No differences were noticed in cell recovery and viability between the groups. Fluorescence-activated cell-sorting (FACS) analyzes on freshly isolated cells indicated that the 1.073 g/mL GMP DGM more efficiently depleted the CD45(+) fraction in comparison with 1.077 GMP DGM. Moreover, in the 1.073 group, fibroblastic colony-forming units (CFU-F) were 1.5 times higher and the final MSC yield 1.8 times increased after four passages. Both reagents isolated MSC with the expected phenotype; however, 1.073-isolated MSC showed a higher expression of CD90, CD146 and GD2. Additionally, MSC from both groups were capable of fully differentiating into bone, adipose cells and cartilage. CONCLUSIONS Both GMP DGM enriched MSC from BM samples, suggesting that these reagents would be suitable for clinical-grade expansions. In addition, the density of 1.073 g/mL provides a significant advantage over 1.077 g/mL GMP DGM, impacting the quantity of MSC obtained and reducing the ex vivo expansion time for optimized cell-based clinical applications.


BioMed Research International | 2013

Isolation, Characterization, and Transduction of Endometrial Decidual Tissue Multipotent Mesenchymal Stromal/Stem Cells from Menstrual Blood

Filippo Rossignoli; Anna Caselli; Giulia Grisendi; Serena Piccinno; Jorge S. Burns; Alba Murgia; Elena Veronesi; Pietro Loschi; Cristina Masini; Pierfranco Conte; Paolo Paolucci; Edwin M. Horwiz; Massimo Dominici

Mesenchymal stromal/stem cells (MSCs) reveal progenitor cells-like features including proliferation and differentiation capacities. One of the most historically recognized sources of MSC has been the bone marrow, while other sources recently include adipose tissue, teeth, bone, muscle, placenta, liver, pancreas, umbilical cord, and cord blood. Frequently, progenitor isolation requires traumatic procedures that are poorly feasible and associated with patient discomfort. In the attempt to identify a more approachable MSC source, we focused on endometrial decidual tissue (EDT) found within menstrual blood. Based also on recent literature findings, we hypothesized that EDT may contain heterogeneous populations including some having MSC-like features. Thus, we here sought to isolate EDT-MSC processing menstrual samples from multiple donors. Cytofluorimetric analyses revealed that resulting adherent cells were expressing mesenchymal surface markers, including CD56, CD73, CD90, CD105 and CD146, and pluripotency markers such as SSEA-4. Moreover, EDT-MSC showed a robust clonogenic potential and could be largely expanded in vitro as fibroblastoid elements. In addition, differentiation assays drove these cells towards osteogenic, adipogenic, and chondrogenic lineages. Finally, for the first time, we were able to gene modify these progenitors by a retroviral vector carrying the green fluorescent protein. From these data, we suggest that EDT-MSC could represent a new promising tool having potential within cell and gene therapy applications.


British Journal of Haematology | 2012

In vitro anti-myeloma activity of TRAIL-expressing adipose-derived mesenchymal stem cells.

Sabino Ciavarella; Giulia Grisendi; Massimo Dominici; Marco Tucci; Oronzo Brunetti; Franco Dammacco; Franco Silvestris

Recently, genetically modified mesenchymal stem cells (MSCs) have been exploited to deliver anti‐cancer bio‐drugs directly within the tumour mass. Here, we explored whether adipose‐derived MSCs (AD‐MSCs), engineered to express the pro‐apoptotic ligand TRAIL (also known as TNFSF10), kill multiple myeloma (MM) cells and migrate towards MM cells in vitro. Different MM cell lines were assessed for their sensitivity to recombinant human (rh) TRAIL alone and in combination with the proteasome inhibitor bortezomib, which was shown to enhance the effect of rhTRAIL. TRAIL+‐AD‐MSCs were co‐cultured with bortezomib‐pretreated MM cells and their killing activity was evaluated in presence or absence of caspase inhibition. AD‐MSC migration towards media conditioned by both myeloma cells and myeloma bone fragments was also investigated. Despite moderate MM cell sensitivity to rhTRAIL, TRAIL+‐AD‐MSCs in combination with bortezomib significantly induced myeloma cell death. This effect was associated with caspase‐8 activation and abrogated by capsase inhibition. On the other hand, co‐culture experiments were performed to evaluate whether unmodified AD‐MSCs affect myeloma cell growth in vitro. AD‐MSCs appeared ineffective on myeloma cell growth and showed migratory capacity towards MM cells in vitro. These data emphasize the anti‐myeloma activity of TRAIL‐engineered AD‐MSCs and provide support for a future model of a cell‐based approach against MM.


Advances in Biochemical Engineering \/ Biotechnology | 2012

MSC and Tumors: Homing, Differentiation, and Secretion Influence Therapeutic Potential

Naomi D’souza; Jorge S. Burns; Giulia Grisendi; Olivia Candini; Elena Veronesi; Serena Piccinno; Edwin M. Horwitz; Paolo Paolucci; Pierfranco Conte; Massimo Dominici

: Mesenchymal stromal/stem cells (MSC) are adult multipotent progenitors with fibroblast-like morphology able to differentiate into adipocytic, osteogenic, chondrogenic, and myogenic lineages. Due to these properties, MSC have been studied and introduced as therapeutics in regenerative medicine. Preliminary studies have also shown a possible involvement of MSC as precursors of cellular elements within tumor microenvironments, in particular tumor-associated fibroblasts (TAF). Among a number of different possible origins, TAF may originate from a pool of circulating progenitors from bone marrow or adipose tissue-derived MSC. There is growing evidence to corroborate that cells immunophenotypically defined as MSC are able to reside as TAF influencing the tumor microenvironment in a potentially bi-phasic and obscure manner: either promoting or inhibiting growth depending on tumor context and MSC sources. Here we focus on relationships between the tumor microenvironment, cancer cells, and MSC, analyzing their diverse ability to influence neoplastic development. Associated activities include MSC homing driven by the secretion of various mediators, differentiation towards TAF phenotypes, and reciprocal interactions with the tumor cells. These are reviewed here with the aim of understanding the biological functions of MSC that can be exploited for innovative cancer therapy.


Stem Cells | 2013

IGF‐1‐mediated osteoblastic niche expansion enhances long‐term hematopoietic stem cell engraftment after murine bone marrow transplantation

Anna Caselli; Timothy S. Olson; Satoru Otsuru; Xiaohua Chen; Ted J. Hofmann; Hyun-Duck Nah; Giulia Grisendi; Paolo Paolucci; Massimo Dominici; Edwin M. Horwitz

The efficiency of hematopoietic stem cell (HSC) engraftment after bone marrow (BM) transplantation depends largely on the capacity of the marrow microenvironment to accept the transplanted cells. While radioablation of BM damages osteoblastic stem cell niches, little is known about their restoration and mechanisms governing their receptivity to engraft transplanted HSCs. We previously reported rapid restoration and profound expansion of the marrow endosteal microenvironment in response to marrow radioablation. Here, we show that this reorganization represents proliferation of mature endosteal osteoblasts which seem to arise from a small subset of high‐proliferative, relatively radio‐resistant endosteal cells. Multiple layers of osteoblasts form along the endosteal surface within 48 hours after total body irradiation, concomitant with a peak in marrow cytokine expression. This niche reorganization fosters homing of the transplanted hematopoietic cells to the host marrow space and engraftment of long‐term‐HSC. Inhibition of insulin‐like growth factor (IGF)‐1‐receptor tyrosine kinase signaling abrogates endosteal osteoblast proliferation and donor HSC engraftment, suggesting that the cytokine IGF‐1 is a crucial mediator of endosteal niche reorganization and consequently donor HSC engraftment. Further understanding of this novel mechanism of IGF‐1‐dependent osteoblastic niche expansion and HSC engraftment may yield clinical applications for improving engraftment efficiency after clinical HSC transplantation. Stem Cells 2013;31:2193–2204


Neoplasia | 2014

Suppression of invasion and metastasis of triple-negative breast cancer lines by pharmacological or genetic inhibition of slug activity.

Giovanna Ferrari-Amorotti; Claudia Chiodoni; Fei Shen; Sara Cattelani; Angela Rachele Soliera; Gloria Manzotti; Giulia Grisendi; Massimo Dominici; Francesco Rivasi; Mario P. Colombo; Alessandro Fatatis; Bruno Calabretta

Most triple-negative breast cancers (TNBCs) exhibit gene expression patterns associated with epithelial-to-mesenchymal transition (EMT), a feature that correlates with a propensity for metastatic spread. Overexpression of the EMT regulator Slug is detected in basal and mesenchymal-type TNBCs and is associated with reduced E-cadherin expression and aggressive disease. The effects of Slug depend, in part, on the interaction of its N-terminal SNAG repressor domain with the chromatin-modifying protein lysine demethylase 1 (LSD1); thus, we investigated whether tranylcypromine [also known as trans-2-phenylcyclopropylamine hydrochloride (PCPA) or Parnate], an inhibitor of LSD1 that blocks its interaction with Slug, suppresses the migration, invasion, and metastatic spread of TNBC cell lines. We show here that PCPA treatment induces the expression of E-cadherin and other epithelial markers and markedly suppresses migration and invasion of TNBC cell lines MDA-MB-231 and BT-549. These effects were phenocopied by Slug or LSD1 silencing. In two models of orthotopic breast cancer, PCPA treatment reduced local tumor growth and the number of lung metastases. In mice injected directly in the blood circulation with MDA-MB-231 cells, PCPA treatment or Slug silencing markedly inhibited bone metastases but had no effect on lung infiltration. Thus, blocking Slug activity may suppress the metastatic spread of TNBC and, perhaps, specifically inhibit homing/colonization to the bone.


Clinical Cancer Research | 2017

Blocking tumor-educated MSC paracrine activity halts osteosarcoma progression

S. Rubina Baglio; Tonny Lagerweij; Maria Pérez-Lanzón; Xuan Dung Ho; Nicolas Léveillé; Sonia A. Melo; Anne-Marie Cleton-Jansen; Ekaterina S. Jordanova; Laura Roncuzzi; Michelina Greco; Monique van Eijndhoven; Giulia Grisendi; Massimo Dominici; Roberta Bonafede; Sinéad M. Lougheed; Tanja D. de Gruijl; Nicoletta Zini; Silvia Cervo; Agostino Steffan; Vincenzo Canzonieri; Aare Märtson; Katre Maasalu; Sulev Kõks; Tom Wurdinger; Nicola Baldini; D. Michiel Pegtel

Purpose: Human osteosarcoma is a genetically heterogeneous bone malignancy with poor prognosis despite the employment of aggressive chemotherapy regimens. Because druggable driver mutations have not been established, dissecting the interactions between osteosarcoma cells and supporting stroma may provide insights into novel therapeutic targets. Experimental Design: By using a bioluminescent orthotopic xenograft mouse model of osteosarcoma, we evaluated the effect of tumor extracellular vesicle (EV)–educated mesenchymal stem cells (TEMSC) on osteosarcoma progression. Characterization and functional studies were designed to assess the mechanisms underlying MSC education. Independent series of tissue specimens were analyzed to corroborate the preclinical findings, and the composition of patient serum EVs was analyzed after isolation with size-exclusion chromatography. Results: We show that EVs secreted by highly malignant osteosarcoma cells selectively incorporate a membrane-associated form of TGFβ, which induces proinflammatory IL6 production by MSCs. TEMSCs promote tumor growth, accompanied with intratumor STAT3 activation and lung metastasis formation, which was not observed with control MSCs. Importantly, intravenous administration of the anti-IL6 receptor antibody tocilizumab abrogated the tumor-promoting effects of TEMSCs. RNA-seq analysis of human osteosarcoma tissues revealed a distinct TGFβ-induced prometastatic gene signature. Tissue microarray immunostaining indicated active STAT3 signaling in human osteosarcoma, consistent with the observations in TEMSC-treated mice. Finally, we isolated pure populations of EVs from serum and demonstrated that circulating levels of EV-associated TGFβ are increased in osteosarcoma patients. Conclusions: Collectively, our findings suggest that TEMSCs promote osteosarcoma progression and provide the basis for testing IL6- and TGFβ-blocking agents as new therapeutic options for osteosarcoma patients. Clin Cancer Res; 23(14); 3721–33. ©2017 AACR.

Collaboration


Dive into the Giulia Grisendi's collaboration.

Top Co-Authors

Avatar

Massimo Dominici

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Paolo Paolucci

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Edwin M. Horwitz

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Elena Veronesi

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlotta Spano

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Filippo Rossignoli

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Olivia Candini

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Giulia Golinelli

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Rita Bussolari

University of Modena and Reggio Emilia

View shared research outputs
Researchain Logo
Decentralizing Knowledge