Network


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

Hotspot


Dive into the research topics where Nicoletta Eliopoulos is active.

Publication


Featured researches published by Nicoletta Eliopoulos.


Stem Cells | 2003

Hypoxia Promotes Murine Bone-Marrow-Derived Stromal Cell Migration and Tube Formation

Borhane Annabi; Ying-Ta Lee; Sandra Turcotte; Emmanuelle Naud; Richard R. Desrosiers; Martin Champagne; Nicoletta Eliopoulos; Jacques Galipeau; Richard Béliveau

Recent evidence indicates that bone‐marrow‐derived stromal cells (MSCs) have a histology coherent with endothelial cells that may enable them to contribute to tumor angiogenesis through yet undefined mechanisms. In this work, we investigated the angiogenic properties of murine MSCs involved in extracellular matrix degradation and in neovascularization that could take place in a hypoxic environment such as that encountered in tumor masses. MSCs were cultured in normoxia (95% air and 5% CO2) or in hypoxia (1% oxygen, 5% CO2, and 94% nitrogen). We found that hypoxic culture conditions rapidly induced MSC migration and three‐dimensional capillary‐like structure formation on Matrigel. In vitro, MSC migration was induced by growth‐factor‐ and cytokine‐enriched conditioned media isolated from U‐87 glioma cells as well as from MSCs cultured in hypoxic conditions, suggesting both paracrine and autocrine regulatory mechanisms. Although greater vascular endothelial growth factor levels were secreted by MSCs in hypoxic conditions, this growth factor alone could not explain their greater migration. Interestingly, matrix metalloproteinase (MMP)‐2 mRNA expression and protein secretion were downregulated, while those of membrane‐type (MT)1‐MMP were strongly induced by hypoxia. Functional inhibition of MT1‐MMP by a blocking antibody strongly suppressed MSC ability to migrate and generate capillary‐like structures. Collectively, these data suggest that MSCs may have the capacity to participate in tumor angiogenesis through regulation of their angiogenic properties under an atmosphere of low oxygen that closely approximates the tumor microenvironment.


Journal of Cellular Biochemistry | 2004

Vascular progenitors derived from murine bone marrow stromal cells are regulated by fibroblast growth factor and are avidly recruited by vascularizing tumors

Borhane Annabi; Emmanuelle Naud; Ying-Ta Lee; Nicoletta Eliopoulos; Jacques Galipeau

Bone marrow‐derived stromal cells (BMSC) possess a population of vascular progenitor cells that enable them to acquire a histology and immunophenotype coherent with endothelial cells (EC). Recent evidence indicates that a hypoxic environment such as that encountered in tumor masses regulates BMSC angiogenic properties by pathways that remain to be defined. It is also unclear as to what extent these marrow‐derived precursor cells could contribute to the growth of endothelium‐lined vessels at the vicinity of tumor masses. In this study, we found that BMSC exhibited the ability to generate three‐dimensional capillary‐like networks on Matrigel, and that this property was up‐regulated by growth factors‐enriched conditioned media isolated from several tumor‐derived cell lines. In particular, basic fibroblast growth factor, a key mediator of angiogenesis, was found to be the most potent growth factor for inducing BMSC proliferation, migration, and tubulogenesis. The setup of a new two‐dimensional in vitro co‐culture assay further showed that BMSC were massively recruited when cultured in the presence of either cancerous or differentiated EC lines. In vivo, subcutaneous co‐injection of BMSC with U‐87 glioma cells in nude mice resulted in the formation of highly vascularized tumors, where BMSC differentiated into CD31‐positive cells and localized at the lumen of vascular structures. Our data suggest that BMSC could be recruited at the sites of active tumor neovascularization through paracrine regulation of their angiogenic properties. These observations may have crucial implications in the development of novel therapies using BMSC engineered to secrete anti‐cancerous agents and to antagonize tumor progression.


Experimental Hematology | 2003

Matrix metalloproteinase regulation of sphingosine-1-phosphate-induced angiogenic properties of bone marrow stromal cells

Borhane Annabi; Sébastien Thibeault; Ying-Ta Lee; Nathalie Bousquet-Gagnon; Nicoletta Eliopoulos; Stéphane Barrette; Jacques Galipeau; Richard Béliveau

OBJECTIVE Bone marrow-derived stromal cells (MSC) are able to acquire histological and immunophenotypic characteristics consistent with endothelial cells (EC). In this study we examined the effect of sphingosine-1-phosphate (S1P), a platelet-derived bioactive lysophospholipid that is believed to specifically stimulate EC migration and tube formation, on the angiogenic properties of MSC. METHODS MSC were isolated from murine bone marrow and cultured in the presence of diverse angiogenic growth factors. Using a chemotaxis chamber and Matrigel tubulogenesis assay, we measured the extent of MSC migration and capillary-like structure formation. Western blots and zymography were used to assess the levels and activation states of soluble and membrane-bound matrix metalloproteinase (MMP). RESULTS We found that S1P strongly induced MSC migration and in vitro capillary-like structure formation. Ilomastat, a broad-spectrum MMP inhibitor, antagonized several angiogenic and S1P-mediated events in MSC. These included 1) the inhibition of S1P-induced tube formation, 2) the inhibition of concanavalin-A (Con-A)-mediated proMMP-2 activation, and 3) the inhibition of S1P- and Con-A-induced caspase-3 activity. Moreover, S1P induced membrane type-1 (MT1)-MMP mRNA and protein expression, but paradoxically antagonized its cell surface proteolytic processing. In addition, anti-angiogenic agents such as Ilomastat, Neovastat, and green tea polyphenol epigallocatechin-3-gallate antagonized the S1P-induced migration of MSC as well as that of transfected COS-7 cells overexpressing the recombinant receptor for S1P, EDG-1. CONCLUSION Collectively, our results indicate a crucial role for S1P/EDG-1-mediated angiogenic and survival events in the regulation of microvascular network remodeling by MSC, and may provide a new molecular link between hemostasis and angiogenesis processes.


Journal of The American Society of Nephrology | 2006

Erythropoietin Delivery by Genetically Engineered Bone Marrow Stromal Cells for Correction of Anemia in Mice with Chronic Renal Failure

Nicoletta Eliopoulos; Raymonde F. Gagnon; Moïra François; Jacques Galipeau

The goal of this research was to develop a strategy to couple stem cell and gene therapy for in vivo delivery of erythropoietin (Epo) for treatment of anemia of ESRD. It was shown previously that autologous bone marrow stromal cells (MSCs) can be genetically engineered to secrete pharmacologic amounts of Epo in normal mice. Therefore, whether anemia in mice with mild to moderate chronic renal failure (CRF) can be improved with Epo gene-modified MSCs (Epo+MSCs) within a subcutaneous implant was examined. A cohort of C57BL/6 mice were rendered anemic by right kidney electrocoagulation and left nephrectomy. In these CRF mice, the hematocrit (Hct) dropped from a prenephrectomy baseline of approximately 55% to 40% after induction of renal failure. MSCs from C57BL/6 donor mice were genetically engineered to secrete murine Epo at a rate of 3 to 4 units of Epo/10(6) cells per 24 h, embedded in a collagen-based matrix, and implanted subcutaneously in anemic CRF mice. It was observed that Hct increased after administration of Epo+MSCs, according to cell dose. Implants of 3 million Epo+MSCs per mouse had no effect on Hct, whereas 10 million led to a supraphysiologic effect. The Hct of CRF mice that received 4.5 or 7.5 million Epo+MSCs rose to a peak 54+/-4.0 or 63+/-5.5%, respectively, at 3 wk after implantation and remained above 48 or 54% for >19 wk. Moreover, mice that had CRF and received Epo+MSCs showed significantly greater swimming exercise capacity. In conclusion, these results demonstrate that subcutaneous implantation of Epo-secreting genetically engineered MSCs can correct anemia that occurs in a murine model of CRF.


Cancer Research | 2008

Neo-Organoid of Marrow Mesenchymal Stromal Cells Secreting Interleukin-12 for Breast Cancer Therapy

Nicoletta Eliopoulos; Moïra François; Marie-Noëlle Boivin; Daniel Martineau; Jacques Galipeau

Bone marrow-derived mesenchymal stromal cells (MSCs), beneficial for regenerative medicine applications due to their wide differentiation capabilities, also hold promise as cellular vehicles for the delivery of therapeutic plasma-soluble gene products due to their ease of handling, expansion, and genetic engineering. We hypothesized that MSCs, gene enhanced to express interleukin-12 (IL-12) and then embedded in a matrix, may act as an anticancer neo-organoid when delivered s.c. in autologous/syngeneic hosts. We performed such experiments in mice and noted that primary murine MSCs retrovirally engineered to secrete murine IL-12 can significantly interfere with growth of 4T1 breast cancer cells in vivo, with a more substantial anticancer action achieved when these cells are embedded in a matrix. Plasma of mice that received the IL-12 MSC-containing neo-organoids showed increased levels of IL-12 and IFN-gamma. Histopathologic analysis revealed less tumor cells in implants of 4T1 cells with IL-12 MSCs, and the presence of necrotic tumor islets and necrotic capillaries, suggesting antiangiogenesis. We also showed that the anticancer effect exerted by the IL-12 MSCs is immune mediated because it is absent in immunodeficient mice, is not due to systemic IL-12 delivery, and also occurs in a B16 melanoma model. This study therefore establishes the feasibility of using gene-enhanced MSCs in a cell-based neo-organoid approach for cancer treatment.


American Journal of Physiology-renal Physiology | 2010

Human marrow-derived mesenchymal stromal cells decrease cisplatin renotoxicity in vitro and in vivo and enhance survival of mice post-intraperitoneal injection

Nicoletta Eliopoulos; Jing Zhao; Manaf Bouchentouf; Kathy Forner; Elena Birman; Shala Yuan; Marie-Noëlle Boivin; Daniel Martineau

Acute kidney injury (AKI) can occur from the toxic side-effects of chemotherapeutic agents such as cisplatin. Bone marrow-derived mesenchymal stromal cells (MSCs) have demonstrated wide therapeutic potential often due to beneficial factors they secrete. The goal of this investigation was to evaluate in vitro the effect of human MSCs (hMSCs) secretome on cisplatin-treated human kidney cells, and in vivo the consequence of hMSCs intraperitoneal (ip) implantation in mice with AKI. Our results revealed that hMSCs-conditioned media improved survival of HK-2 human proximal tubular cells exposed to cisplatin in vitro. This enhanced survival was linked to increased expression of phosphorylated Akt (Ser473) and was reduced by a VEGF-neutralizing antibody. In vivo testing of these hMSCs established that ip administration in NOD-SCID mice decreased cisplatin-induced kidney function impairment, as demonstrated by lower blood urea nitrogen levels and higher survival. In addition, blood phosphorous and amylase levels were also significantly decreased. Moreover, hMSCs reduced the plasma levels of several inflammatory cytokines/chemokines. Immunohistochemical examination of kidneys showed less apoptotic and more proliferating cells. Furthermore, PCR indicated the presence of hMSCs in mouse kidneys, which also showed enhanced expression of phosphorylated Akt. In conclusion, our study reveals that hMSCs can exert prosurvival effects on renal cells in vitro and in vivo, suggests a paracrine contribution for kidney protective abilities of hMSCs delivered ip, and supports their clinical potential in AKI.


Cardiovascular Research | 2008

Coupling erythropoietin secretion to mesenchymal stromal cells enhances their regenerative properties

Ian B. Copland; E. Marc Jolicoeur; Marc-Antoine Gillis; Jessica Cuerquis; Nicoletta Eliopoulos; Borhane Annabi; Angelo Calderone; Jean-François Tanguay; Anique Ducharme; Jacques Galipeau

AIMS Mesenchymal stromal cells (MSCs) possess intrinsic features that identify them as useful for treating ischaemic syndromes. Poor in vivo survival/engraftment of MSCs, however, limits their overall effectiveness. In this work, we tested whether genetically engineering MSCs to secrete erythropoietin (Epo) could represent a better therapeutic platform than MSCs in their native form. METHODS AND RESULTS MSCs from C57Bl/6 mice were retrovirally transduced with either an empty vector or one that causes the production of Epo and were then analysed for the alterations in angiogenic and survival potential. Using a mouse model of myocardial infarction (MI), the regenerative potential of null MSCs and Epo-overexpressing MSCs (Epo+MSCs) was assessed using serial echocardiogram and invasive haemodynamic measurements. Infarct size, capillary density and neutrophil influx were assessed using histologic techniques. Using in vitro assays coupled with an in vivo Matrigel plug assay, we demonstrate that engineering MSCs to express Epo does not alter their immunophenotype or plasticity. However, relative to mock-modified MSCs [wild-type (WT)-MSCs], Epo+MSCs are more resilient to apoptotic stimuli and initiate a more robust host-derived angiogenic response. We also identify and characterize the autocrine loop established on MSCs by having them secrete Epo. Furthermore, in a murine model of MI, animals receiving intracardiac injections of Epo+MSCs exhibited significantly enhanced cardiac function compared with WT-MSCs and saline-injected control animals post-MI, owing to the increased myocardial capillary density and the reduced neutrophilia. CONCLUSION Epo overexpression enhances the cellular regenerative properties of MSCs by both autocrine and paracrine pathways.


Human Molecular Genetics | 2011

A variant OSR1 allele which disturbs OSR1 mRNA expression in renal progenitor cells is associated with reduction of newborn kidney size and function

Zhao Zhang; Diana M. Iglesias; Nicoletta Eliopoulos; Reyhan El Kares; LeeLee Chu; Paola Romagnani; Paul Goodyer

Human nephrons are formed during fetal life through an interaction between the branching ureteric bud and progenitor cells. The wide variation in final nephron number has been attributed to allelic variants of genes regulating ureteric bud arborization. Here, we hypothesize that dysfunctional variants of the Odd-Skipped Related 1 (OSR1) gene which compromise the renal progenitor cell pool might also limit newborn kidney size and function. We show that OSR1 is expressed in human mesenchymal stem cells, the blastemal component of Wilms tumors and CD24+/CD133+ progenitor cells isolated from the mature kidney. We identified an OSR1(rs12329305(T)) allele in 6% of normal Caucasians which alters an exon2 splice enhancer. This variant is predicted to reduce spliceosome-binding affinity and stability of the OSR1 mRNA. In cultured cells, the OSR1(rs12329305)(T) allele produced no identifiable transcript. Normal Caucasian newborns from Montreal with the OSR1(rs12329305)(T) allele had kidney volume 11.8% smaller (P= 0.006) and cord blood cystatin C levels 12.6% higher (P = 0.005) than those with wild-type genotype. Effects of the OSR1(rs12329305)(T) allele are additive with genes that alter ureteric bud branching. Kidney volume was reduced more in newborns bearing both RET(rs1800860)(A) and OSR1(rs12329305)(T) alleles (22%, P= 0.0008) and cystatin C was increased by 17% (P= 0.006) versus newborns with wild-type alleles. Although only two subjects had PAX2(rs11599825)(A) and OSR1(rs12329305)(T) alleles, kidney size was reduced by 27% and cystatin C was increased by 14% versus wild-types (P= NS).


Cytotherapy | 2016

Adipose mesenchymal stromal cells response to ionizing radiation.

Osama Muhammad Maria; Slawomir Kumala; Mitra Heravi; Alasdair Syme; Nicoletta Eliopoulos; Thierry Muanza

BACKGROUND AIMS This study evaluates the biological response of adipose tissue-derived mesenchymal stromal cells (aMSCs) to ionizing radiation (IR). METHODS Irradiated BALB/c mice aMSCs were characterized for functionality and phenotype. The clonogenic capacity of irradiated aMSCs was assessed and compared with those of metastatic breast cancer cell line (4T1) and normal mouse fibroblasts (NIH3T3-wt). We investigated the IR-induced DNA damage response, apoptosis, changes in cell cycle (CC) dynamics and protein and gene expression. RESULTS Irradiated and non-irradiated aMSCs were able to differentiate into adipocytes, chondrocytes and osteocytes with no significant difference. Irradiated aMSCs maintained the expression of mesenchymal stromal cells (MSCs) surface antigens and, as expected, were negative for hematopoietic stem cells (HSCs) surface antigens when tested up to 7 days after IR for all irradiation doses with no significant difference. Clonogenically, irradiated aMSCs had higher relative survival fraction and plating efficiency than 4T1 and NIH3T3-wt. Irradiated aMSCs expressed higher □H2AX and significantly showed faster and more time-efficient IR-induced DNA damage response evident by up-regulated DNA-PKcs and RAD51. Two hours after IR, most of aMSCs DNA damage/repair-related genes showed up-regulation that disappeared within 6 h after IR. Irradiated aMSCs showed a significant rise and an earlier peak of p-ATM-dependent and -independent (p84/5E10-mediated) G2/M CC arrest compared with 4T1 and NIH3T3-wt. CONCLUSIONS After IR exposure, aMSCs showed a robust and time-efficient radiation-induced DNA damage repair response, stable phenotypical characteristics and multi-lineage differentiation potential, suggesting they may be reliable candidates for cell therapy in radiation oncology regenerative medicine.


Journal of Stem Cell Research and Medicine | 2016

Mesenchymal stromal cells therapy in radiation oncology regenerative medicine

Osama Muhammad Maria; Nicoletta Eliopoulos; Thierry Muanza

Mesenchymal stromal cells (MSCs) are multipotent somatic cells resident in many tissues and organs. They have specific characteristics that distinguish them from other cell types. They are self-renewing cells with multi-lineage differentiation potential. In addition, they possess anti-inflammatory and immunomodulatory properties. Studies have shown that they could be used as vehicles to deliver certain therapeutic gene products as well. These cells possess secretory capabilities of certain cytokines and growth factors that mediate various paracrine effects. They increase the secretion of the anti-inflammatory interleukin-10 (IL-10) together with lowering the availabilities of tumor necrosis factor-alpha (TNF-α), interferon-gamma (INF-γ), and interleukin -1-beta (IL-1β) by signaling to the immune system elements, e.g. dendritic cells, T-cells, B-cells, and natural Killer cells (NK cells). Recently, studies have investigated such anti-inflammatory properties of MSCs in the repair of radiation-induced normal tissue injury, also called radiation oncology regenerative medicine (RORM), supported by the recently known MSCs radiation resistance potential. In this review, we summarize MSCs radio-resistant mechanisms, anti-inflammatory properties, and their application in RORM with special attention to adipose tissue-derived MSCs (aMSCs). Correspondence to: Dr. Thierry Muanza, MD, MSc, FRCPC, Radiation Oncology Translational Research Lab, Department of Radiation Oncology, Jewish General Hospital and Lady Davis Institute Research Centre, McGill University, 3755 Côte-St.-Catherine Road, Suite G002, Montréal, Québec, Canada, H3T 1E2, Tel: +1 (514)-340-8288, Fax: + 1 (514)-340-7548, E-mail: [email protected]

Collaboration


Dive into the Nicoletta Eliopoulos's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Borhane Annabi

Université du Québec à Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ying-Ta Lee

Université du Québec à Montréal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emmanuelle Naud

Université du Québec à Montréal

View shared research outputs
Researchain Logo
Decentralizing Knowledge