Network


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

Hotspot


Dive into the research topics where Lilia Levdansky is active.

Publication


Featured researches published by Lilia Levdansky.


Bone Marrow Transplantation | 2006

Isolation of mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads.

Ibrahim Kassis; Lior Zangi; Rachel Rivkin; Lilia Levdansky; S Samuel; Gerald R. Marx; Raphael Gorodetsky

Adult mesenchymal stem cells (MSC) that are able to differentiate into various mesenchymal cell types are typically isolated from bone marrow, but their significant presence in human peripheral blood (PB) is controversial. Fibrin microbeads (FMB) that bind matrix-dependent cells were used to isolate MSC from the mononuclear fraction of mobilized PB of adult healthy human donors treated with a granulocyte colony-stimulating factor. Isolation by plastic adherence resulted in a negligible number of MSC in all samples tested, whereas FMB-based isolation yielded spindle-shaped cell samples that could further expand on plastic or on FMB in eight out of the 11 samples. The yield of these cells at days 17–18 after the harvest was ∼0.5% of the initial cell number. The isolated cells were grown on plastic and characterized by FACS analysis and immunohistochemistry for specific markers. Following culturing and first passage, the FMB-isolated cells stained positive for mesenchymal stromal cell markers CD90 and CD105, expressed vimentin and fibronectin and were negative for hematopoietic markers CD45 and CD34. These cells could differentiate into osteoblasts, adipocytes and chondrocytes. This study indicates that FMB may have special advantage in isolating MSC from sources such as mobilized PB, where the number of such cells is scarce.


Cloning and Stem Cells | 2007

High-Yield Isolation, Expansion, and Differentiation of Murine Bone Marrow-Derived Mesenchymal Stem Cells Using Fibrin Microbeads (FMB)

Rachel Rivkin; Alon Ben-Ari; Ibrahim Kassis; Lior Zangi; Elena Gaberman; Lilia Levdansky; Gerard Marx; Raphael Gorodetsky

Transplantation of adult mesenchymal stem cells (MSCs) could provide a basis for tissue regeneration. MSCs are typically isolated from bone marrow (BM) based on their preferential adherence to plastic, although with low efficiency in terms of yield and purity. Extensive expansion is needed to reach a significant number of MSCs for any application. Fibrin microbeads (FMB) were designed to attach mesenchymal cells and to provide a matrix for their expansion. The current study was aimed at isolating a high yield of purified BM-derived mouse MSCs based on their preferential adherence and proliferation on FMB in suspension cultures. MSCs could be downloaded to plastics or further expanded on FMB. The yield of MSCs obtained by the FMB isolation technique was about one order of magnitude higher than that achieved by plastic adherence, suggesting that these cells are more abundant than previously reported. FMB-isolated cells were classified as MSCs by their fibroblastic morphology, self-renewal ability, and expression profile of their surface antigens, as examined by flow cytometry and immunostaining. In cell culture, the isolated MSCs could be induced to differentiate into three different mesodermal lineages, as demonstrated by histochemical stains and by RT-PCR analyses of tissue-specific genes. MSCs were also able to differentiate into osteocytes while still cultured on FMB. Our results suggest that FMB might serve as an efficient platform for the isolation, expansion, and differentiation of mouse BM-derived MSCs to be subsequently implanted for tissue regeneration.


Tissue Engineering Part A | 2009

Isolation and implantation of bone marrow-derived mesenchymal stem cells with fibrin micro beads to repair a critical-size bone defect in mice.

Alon Ben-Ari; Rachel Rivkin; Miryam Frishman; Elena Gaberman; Lilia Levdansky; Raphael Gorodetsky

Fibrin microbeads (FMBs) made using thermal treatment of fibrin drops in oil can efficiently isolate mesenchymal stem cells (MSCs) from bone marrow (BM) and other similar sources and culture them continuously in suspension culture. The pure mesenchymal profile of MSCs isolated using FMBs and their differentiation potency to different mesenchymal lineages were previously described in detail. In the current study, MSCs were isolated from the BM of (GFP+) C57/bl mice using FMBs. Addition of pro-osteogenic medium with 10 mM of ss-glycerolphosphate, 50 microg/mL of ascorbic acid, and 10(-8) M of dexamethasone for 1 month resulted in ossified bone-like solid cellular structures, as seen using fluorescence and scanning electron microscopy (SEM). Such spontaneously formed structures were implanted in full-depth approximately 5-mm-diameter drilled defects in the skulls of wild-type c57/bl mice. Two months later, the excised upper parts of the skulls with the defects were viewed using fluorescence microscopy for green fluorescence protein of the cells in the defect and using SEM. They were also scanned using micro-computed tomography to visualize the formation of new hard tissue. Then the samples were processed and sectioned for hematoxylin and eosin staining and immunohistochemistry. Implanted FMBs loaded with (GFP+) MSCs formed partially mature, dense bone-like tissue using a residual moderate inflammatory process containing remnants of FMBs and neo-angiogenesis. The filled defect with bone-like tissue had a Ca/P ratio similar to that of native bone. Limited merging of the implant with the skull indicated that the induced bone regeneration derived from the MSCs that were delivered with the implant. No repair was seen in the control animals without implants or where the defect was filled with FMBs only. Repair scoring (on a 0-5 scale) was found to be 3.38+/-0.35 in the experimental arm, relative to 0 in the controls (p < 0.001).


Nephrology Dialysis Transplantation | 2008

A 3D rotary renal and mesenchymal stem cell culture model unveils cell death mechanisms induced by matrix deficiency and low shear stress

Nilly Shimony; Idit Avrahami; Raphael Gorodetsky; Gregory Elkin; Keren Tzukert; Lior Zangi; Lilia Levdansky; Lina Krasny; Yosef S. Haviv

BACKGROUND In epithelial and endothelial cells, detachment from the matrix results in anoikis, a form of apoptosis, whereas stromal and cancer cells are often anchorage independent. The classical anoikis model is based on static 3D epithelial cell culture conditions (STCK). METHODS We characterized a new model of renal, stromal and mesenchymal stem cell (MSC) matrix deprivation, based on slow rotation cell culture conditions (ROCK). This model induces anoikis using a low shear stress, laminar flow. The mechanism of cell death was determined via FACS (fluorescence-activated cell sorting) analysis for annexin V and propidium iodide uptake and via DNA laddering. RESULTS While only renal epithelial cells progressively died in STCK, the ROCK model could induce apoptosis in stromal and transformed cells; cell survival decreased in ROCK versus STCK to 40%, 52%, 62% and 7% in human fibroblast, rat MSC, renal cell carcinoma (RCC) and human melanoma cell lines, respectively. Furthermore, while ROCK induced primarily apoptosis in renal epithelial cells, necrosis was more prevalent in transformed and cancer cells [necrosis/apoptosis ratio of 72.7% in CaKi-1 RCC cells versus 4.3% in MDCK (Madin-Darby canine kidney) cells]. The ROCK-mediated shift to necrosis in RCC cells was further accentuated 3.4-fold by H(2)O(2)-mediated oxidative stress while in adherent HK-2 renal epithelial cells, oxidative stress enhanced apoptosis. ROCK conditions could also unveil a similar pattern in the LZ100 rat MSC line where in ROCK 44% less apoptosis was observed versus STCK and 45% less apoptosis versus monolayer conditions. Apoptosis in response to oxidative stress was also attenuated in the rat MSC line in ROCK, thereby highlighting rat MSC transformation. CONCLUSIONS The ROCK matrix-deficiency cell culture model may provide a valuable insight into the mechanism of renal and MSC cell death in response to matrix deprivation.


Radiation Research | 1998

Paclitaxel-induced modification of the effects of radiation and alterations in the cell cycle in normal and tumor mammalian cells.

Raphael Gorodetsky; Lilia Levdansky; Israel Ringel; Akiva Vexler

The cytotoxicity of paclitaxel (taxol) is associated mainly with block in G2/M phase, the most radiosensitive phase of the cell cycle. Nevertheless, taxol-induced modification of the effects of radiation may vary from clear sensitization to subadditivity. Therefore, this effect was studied in relation to drug-induced alterations in the distribution of cells in the phases of the cell cycle in tumor cells (EMT-6 and OV-1063) and normal skin fibroblasts. Cell survival was evaluated with two colorimetric assays. The cell cycle was evaluated by FACS analysis of doubly-labeled cells. The radiosensitivity of the different cells studied was similar, apart from the less radiosensitive human fibroblasts. However, their dose- and time-dependent sensitivity to taxol varied significantly. After 24 h exposure of EMT-6 cells to taxol (IC50 approximately 20 nM), the fraction of cells in G2/M phase increased, the fraction in S phase decreased, and the proportion of possibly apoptotic cells with subdiploid and subtetraploid DNA content increased; this coincided with radiosensitization. In OV-1063 cells (IC50 approximately 3 nM), the drug-induced G2/M-phase block was most pronounced, but the combined effect with radiation was merely additive. In human fibroblasts (IC50 approximately 35 nM), a minimal G2/M-phase block with no change in the S phase and a massive elevation of apoptotic cells with subdiploid DNA content was accompanied by a subadditive combined effect with radiation. Six hours of exposure to taxol increased the fraction of cells in S phase in both nonsynchronized and S-phase-synchronized human fibroblasts (G1 phase approximately 65%, S phase approximately 13%). This was accompanied by a pronounced subadditive effect of the combined treatment. However, in G1-phase synchronized human fibroblasts (G1 phase > or =90%, S phase approximately 3%), only the fraction of cells in G2/M phase was slightly elevated, with a merely additive response to the combined treatment. The differences in the response to the combined treatment between slowly and rapidly proliferating cells in relation to modifications in the cell cycle are discussed.


PLOS ONE | 2013

Mitigation of Lethal Radiation Syndrome in Mice by Intramuscular Injection of 3D Cultured Adherent Human Placental Stromal Cells.

Elena Gaberman; Lena Pinzur; Lilia Levdansky; Maria Tsirlin; Nir Netzer; Zami Aberman; Raphael Gorodetsky

Exposure to high lethal dose of ionizing radiation results in acute radiation syndrome with deleterious systemic effects to different organs. A primary target is the highly sensitive bone marrow and the hematopoietic system. In the current study C3H/HeN mice were total body irradiated by 7.7 Gy. Twenty four hrs and 5 days after irradiation 2×106 cells from different preparations of human derived 3D expanded adherent placental stromal cells (PLX) were injected intramuscularly. Treatment with batches consisting of pure maternal cell preparations (PLX-Mat) increased the survival of the irradiated mice from ∼27% to 68% (P<0.001), while cell preparations with a mixture of maternal and fetal derived cells (PLX-RAD) increased the survival to ∼98% (P<0.0001). The dose modifying factor of this treatment for both 50% and 37% survival (DMF50 and DMF37) was∼1.23. Initiation of the more effective treatment with PLX-RAD injection could be delayed for up to 48 hrs after irradiation with similar effect. A delayed treatment by 72 hrs had lower, but still significantly effect (p<0.05). A faster recovery of the BM and improved reconstitution of all blood cell lineages in the PLX-RAD treated mice during the follow-up explains the increased survival of the cells treated irradiated mice. The number of CD45+/SCA1+ hematopoietic progenitor cells within the fast recovering population of nucleated BM cells in the irradiated mice was also elevated in the PLX-RAD treated mice. Our study suggests that IM treatment with PLX-RAD cells may serve as a highly effective “off the shelf” therapy to treat BM failure following total body exposure to high doses of radiation. The results suggest that similar treatments may be beneficial also for clinical conditions associated with severe BM aplasia and pancytopenia.


Journal of Investigative Dermatology | 2013

Marrow-Derived Stromal Cell Delivery on Fibrin Microbeads Can Correct Radiation-Induced Wound-Healing Deficits

Michael W. Xie; Raphael Gorodetsky; Ewa D. Micevicz; Natalia C. Mackenzie; Elena Gaberman; Lilia Levdansky; William H. McBride

Skin that is exposed to radiation has an impaired ability to heal wounds. This is especially true for whole body irradiation, where even moderate non-lethal doses can result in wound healing deficits. Our previous attempts to administer dermal cells locally to wounds to correct radiation-induced deficits were hampered by poor cell retention. Here we improve the outcome by using biodegradable fibrin microbeads (FMB) to isolate a population of mesenchymal marrow-derived stromal cells (MSC) from murine bone marrow by their specific binding to the fibrin matrix, culture them to high density in vitro and deliver them as MSC on FMB at the wound site. MSC are retained and proliferate locally and assist wounds gain tensile strength in whole body irradiated mice with or without additional skin only exposure. MSC-FMB were effective in 2 different mouse strains but were ineffective across a major histocompatability barrier. Remarkably, irradiated mice whose wounds were treated with MSC-FMB showed enhanced hair regrowth suggesting indirect effect on the correction of radiation-induced follicular damage. Further studies showed that additional wound healing benefit could be gained by administration of G-CSF and AMD3100. Collagen strips coated with haptides and MSCs were also highly effective in correcting radiation-induced wound healing deficits.


Regenerative Medicine | 2010

Efficient isolation and chondrogenic differentiation of adult mesenchymal stem cells with fibrin microbeads and micronized collagen sponges

Reut Shainer; Elena Gaberman; Lilia Levdansky; Raphael Gorodetsky

BACKGROUND Mesenchymal stem cells (MSCs) have been demonstrated to potentially undergo chondrogenic differentiation. We propose a new matrix for stem cell-based chondrogenesis using dense fibrin microbeads (FMBs) combined with grounded dehydrothermally crosslinked collagen sponges (micronized collagen). METHODS In this study, MSCs were isolated from bone marrow of transgenic green fluorescent protein C57/Bl mice by FMBs in high yield. After 48 h in slowly rotating suspension culture, micronized collagen was added. RESULTS The cells on the FMBs migrated to the collagen pieces and formed aggregates that developed into cartilage-like structures. Following chondrogenic differentiation, alcian blue staining and collagen type II immunohistochemistry demonstrated the presence of chondrocytes in the 3D structures. PCR for the expression of aggrecan and collagen type II genes supported these findings. The in vitro structures that formed were used for ectopic subdermal implantation in wild-type C57/Bl mice. However, the chondrogenic markers faded relative to the pre-implant in vitro structures. CONCLUSION We propose that FMBs with micronized collagen could serve as a simple technology for MSC isolation and chondrogenesis as a basis for implantation.


Acta Haematologica | 2007

Effect of Halofuginone, a Collagen α1(I) Inhibitor, on Wound Healing in Normal and Irradiated Skin: Implication for Hematopoietic Stem Cell Transplantation

Arnon Nagler; Meir Ohana; Merav Leiba; Lilia Levdansky; Raphael Gorodetsky

It is a major interest in the field of hematopoietic stem cell transplantation to reduce scarring of healing wounds with overdeposition of collagen due to radiation injury or graft-versus-host disease. Halofuginone (HF) inhibits collagen α1(I) gene expression and overdeposition of collagen. We examined the effect of HF on the healing of full-depth incision wounds inflicted in normal skin or skin areas compromised by local preirradiation with 18 Gy. Preirradiation significantly decreased the tensile strength of the healing wounds at day 14 (by ∼60%, p < 0.0001). In contrast, HF treatment did not significantly decrease the strength of wounds inflicted in both normal and preirradiated skin. Histological evaluation revealed that HF induced moderate thinning of the dermis accompanied by elevated thickness of the epidermis and enhanced rejoining of subdermal muscles in the wound area. HF only minimally reduced total collagen deposition in both groups, with minor changes in the level of more matured fibrillar collagen network. Our study demonstrates that HF does not significantly affect wound strength. This encourages the possible use of HF as an antifibrotic agent with minimal complications for post-hematopoietic stem cell transplantation complications including radiation toxicity and graft-versus-host disease.


Peptides | 2011

Blood pressure modulation following activation of mast cells by cationic cell penetrating peptides

Maamoun Basheer; Herzl Schwalb; Irit Shefler; Lilia Levdansky; Yoseph A. Mekori; Raphael Gorodetsky

Short cell penetrating peptides (CPP) are widely used in vitro to transduce agents into cells. But their systemic effect has not been yet studied in detail. We studied the systemic effect of the cell penetrating peptides, penetratin, transportan and pro-rich, on rat hemodynamic functions. Intra-arterial monitoring of blood pressure showed that injection of the positively charged penetratin and transportan in a wide range of concentrations (2.5-320 μg/kg) caused highly significant transient decrease in the systolic and diastolic blood pressure in a dose dependent manner (p<0.01). Pretreatment with histamine receptors blockers or with cromolyn, a mast cell stabilizing agent, significantly attenuated this effect. Furthermore, in vitro incubation of these both peptides with mast cells line, LAD2, caused a massive mast cell degranulation. In vitro studies showed that these CPP in a wide range of concentrations were not cytotoxic without any effect on the survival of LAD2 mast cell line. In contrast, the less positively charged and proline-rich CPP, pro-rich, had no systemic effects with no effect on mast cell degranulation. Our results indicate that intravenously administrated positively charged CPP may have deleterious consequences due to their induced BP drop, mediated by mast cell activation. Therefore, the major effect of mast cell activation on BP should be considered in developing possible future drug therapies based on the injection of membrane-permeable and positively charged CPP. Nevertheless, lower levels of such CPP may be considered as a treatment of systemic high BP through moderate systemic mast cell activation.

Collaboration


Dive into the Lilia Levdansky's collaboration.

Top Co-Authors

Avatar

Raphael Gorodetsky

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Elena Gaberman

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lior Zangi

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Akiva Vexler

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Assaf Friedler

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Avraham Reshef

Hebrew University of Jerusalem

View shared research outputs
Researchain Logo
Decentralizing Knowledge