Network


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

Hotspot


Dive into the research topics where Yaron Vagima is active.

Publication


Featured researches published by Yaron Vagima.


Blood | 2012

S1P promotes murine progenitor cell egress and mobilization via S1P1-mediated ROS signaling and SDF-1 release

Karin Golan; Yaron Vagima; Aya Ludin; Tomer Itkin; Shiri Cohen-Gur; Alexander Kalinkovich; Orit Kollet; Chihwa Kim; Amir Schajnovitz; Kfir Lapid; Shoham Shivtiel; Andrew J. Morris; Mariusz Z. Ratajczak; Tsvee Lapidot

The mechanisms of hematopoietic progenitor cell egress and clinical mobilization are not fully understood. Herein, we report that in vivo desensitization of Sphingosine-1-phosphate (S1P) receptors by FTY720 as well as disruption of S1P gradient toward the blood, reduced steady state egress of immature progenitors and primitive Sca-1(+)/c-Kit(+)/Lin(-) (SKL) cells via inhibition of SDF-1 release. Administration of AMD3100 or G-CSF to mice with deficiencies in either S1P production or its receptor S1P(1), or pretreated with FTY720, also resulted in reduced stem and progenitor cell mobilization. Mice injected with AMD3100 or G-CSF demonstrated transient increased S1P levels in the blood mediated via mTOR signaling, as well as an elevated rate of immature c-Kit(+)/Lin(-) cells expressing surface S1P(1) in the bone marrow (BM). Importantly, we found that S1P induced SDF-1 secretion from BM stromal cells including Nestin(+) mesenchymal stem cells via reactive oxygen species (ROS) signaling. Moreover, elevated ROS production by hematopoietic progenitor cells is also regulated by S1P. Our findings reveal that the S1P/S1P(1) axis regulates progenitor cell egress and mobilization via activation of ROS signaling on both hematopoietic progenitors and BM stromal cells, and SDF-1 release. The dynamic cross-talk between S1P and SDF-1 integrates BM stromal cells and hematopoeitic progenitor cell motility.


Blood | 2011

Enhanced c-Met activity promotes G-CSF–induced mobilization of hematopoietic progenitor cells via ROS signaling

Melania Tesio; Karin Golan; Simona Corso; Silvia Giordano; Amir Schajnovitz; Yaron Vagima; Shoham Shivtiel; Alexander Kalinkovich; Luisa Caione; Loretta Gammaitoni; Elisa Laurenti; Eike C. Buss; Elias Shezen; Tomer Itkin; Orit Kollet; Isabelle Petit; Andreas Trumpp; James G. Christensen; Massimo Aglietta; Wanda Piacibello; Tsvee Lapidot

Mechanisms governing stress-induced hematopoietic progenitor cell mobilization are not fully deciphered. We report that during granulocyte colony-stimulating factor-induced mobilization c-Met expression and signaling are up-regulated on immature bone marrow progenitors. Interestingly, stromal cell-derived factor 1/CXC chemokine receptor-4 signaling induced hepatocyte growth factor production and c-Met activation. We found that c-Met inhibition reduced mobilization of both immature progenitors and the more primitive Sca-1(+)/c-Kit(+)/Lin(-) cells and interfered with their enhanced chemotactic migration to stromal cell-derived factor 1. c-Met activation resulted in cellular accumulation of reactive oxygen species by mammalian target of rapamycin inhibition of Forkhead Box, subclass O3a. Blockage of mammalian target of rapamycin inhibition or reactive oxygen species signaling impaired c-Met-mediated mobilization. Our data show dynamic c-Met expression and function in the bone marrow and show that enhanced c-Met signaling is crucial to facilitate stress-induced mobilization of progenitor cells as part of host defense and repair mechanisms.


Journal of Clinical Investigation | 2009

MT1-MMP and RECK are involved in human CD34+ progenitor cell retention, egress, and mobilization

Yaron Vagima; Abraham Avigdor; Polina Goichberg; Shoham Shivtiel; Melania Tesio; Alexander Kalinkovich; Karin Golan; Ayelet Dar; Orit Kollet; Isabelle Petit; Orly Perl; Ester Rosenthal; Igor B. Resnick; Izhar Hardan; Yechiel N. Gellman; David Naor; Arnon Nagler; Tsvee Lapidot

The mechanisms governing hematopoietic progenitor cell mobilization are not fully understood. We report higher membrane type 1-MMP (MT1-MMP) and lower expression of the MT1-MMP inhibitor, reversion-inducing cysteine-rich protein with Kazal motifs (RECK), on isolated circulating human CD34+ progenitor cells compared with immature BM cells. The expression of MT1-MMP correlated with clinical mobilization of CD34+ cells in healthy donors and patients with lymphoid malignancies. Treatment with G-CSF further increased MT1-MMP and decreased RECK expression in human and murine hematopoietic cells in a PI3K/Akt-dependent manner, resulting in elevated MT1-MMP activity. Blocking MT1-MMP function by Abs or siRNAs impaired chemotaxis and homing of G-CSF-mobilized human CD34+ progenitors. The mobilization of immature and maturing human progenitors in chimeric NOD/SCID mice by G-CSF was inhibited by anti-MT1-MMP treatment, while RECK neutralization promoted motility and egress of BM CD34+ cells. BM c-kit+ cells from MT1-MMP-deficient mice also exhibited inferior chemotaxis, reduced homing and engraftment capacities, and impaired G-CSF-induced mobilization in murine chimeras. Membranal CD44 cleavage by MT1-MMP was enhanced following G-CSF treatment, reducing CD34+ cell adhesion. Accordingly, CD44-deficient mice had a higher frequency of circulating progenitors. Our results reveal that the motility, adhesion, homing, and mobilization of human hematopoietic progenitor cells are regulated in a cell-autonomous manner by dynamic and opposite changes in MT1-MMP and RECK expression.


Methods of Molecular Biology | 2011

Pathways Implicated in Stem Cell Migration: The SDF-1/CXCR4 Axis

Yaron Vagima; Kfir Lapid; Orit Kollet; Polina Goichberg; Ronen Alon; Tsvee Lapidot

The hallmark of hematopoietic stem and progenitor cells (HSPCs) is their motility, which is essential for their function, such as recruitment upon demand. Stromal Derived Factor-1 (SDF-1, CXCL12) and its major receptor CXCR4 play major roles in stem cell motility and development. In vitro migration assays, implicating either gradients or cell surface-bound forms of SDF-1, are easy to perform and provide vital information regarding directional and random stem cell motility, which correlate with their repopulation potential in clinical and experimental transplantations. In vivo stem cell homing to the bone marrow, their retention, engraftment, and egress to the circulation, all involve SDF-1/CXCR4 interactions. Finally, other stem cell features such as stem cell survival and proliferation, are also dependent on the SDF-1/CXCR4 axis.


Leukemia | 2013

Physiologic corticosterone oscillations regulate murine hematopoietic stem/progenitor cell proliferation and CXCL12 expression by bone marrow stromal progenitors.

Orit Kollet; Yaron Vagima; Gabriele D'Uva; Karin Golan; Jonathan Canaani; Tomer Itkin; Shiri Gur-Cohen; Alexander Kalinkovich; G Caglio; C Medaglia; Aya Ludin; Kfir Lapid; Elias Shezen; A Neufeld-Cohen; D Varol; A Chen; Tsvee Lapidot

The role of corticosterone (Cort), the immune system’s major stress hormone, in the regulation of hematopoietic stem and progenitor cells (HSPCs) and their dynamic bone marrow (BM) microenvironment is currently unknown. We report that corticotropin-releasing factor receptor 1 (CRFR1) mutant mice with chronically low Cort levels showed aberrant HSPC regulation, having higher HSPC numbers and upregulation of the chemokine CXCL12, phenotypes that were restored by Cort supplementation. Expanded stromal progenitors known to support HSPCs were also observed in these low-Cort-containing mice. A similar phenotype was induced in wild-type (WT) mice by Metyrapone, a Cort synthesis inhibitor. Conversely, high Cort exposure induced HSPC apoptosis, reduced long-term BM repopulation and decreased stromal progenitor cell numbers. We documented circadian oscillations of Cort in WT BM but not in CRFR1 mutant mice, leading to diminished circadian BM CXCL12 fluctuations and increased number of circulating HSPCs in these mice. Finally, low Cort induced expansion of stromal progenitors, CXCL12 expression, HSPC proliferation and BM repopulation capacity, involving Notch1 signaling. This was associated with upregulation of the Notch ligand, Jagged1, in BM myeloid cells. Our results suggest that daily physiologic Cort oscillations are critical for balanced HSPC proliferation and function involving Notch1 signaling and their supportive BM microenvironment.


Blood | 2008

Heparanase regulates retention and proliferation of primitive Sca-1+/c-Kit+/Lin− cells via modulation of the bone marrow microenvironment

Asaf Spiegel; Eyal Zcharia; Yaron Vagima; Tomer Itkin; Alexander Kalinkovich; Ayelet Dar; Orit Kollet; Neta Netzer; Karin Golan; Itay Shafat; Neta Ilan; Arnon Nagler; Israel Vlodavsky; Tsvee Lapidot

Heparanase is involved in tumor growth and metastasis. Because of its unique cleavage of heparan sulfate, which binds cytokines, chemokines and proteases, we hypothesized that heparanase is also involved in regulation of early stages of hematopoiesis. We report reduced numbers of maturing leukocytes but elevated levels of undifferentiated Sca-1(+)/c-Kit(+)/Lin(-) cells in the bone marrow (BM) of mice overexpressing heparanase (hpa-Tg). This resulted from increased proliferation and retention of the primitive cells in the BM microenvironment, manifested in increased SDF-1 turnover. Furthermore, heparanase overexpression in mice was accompanied by reduced protease activity of MMP-9, elastase, and cathepsin K, which regulate stem and progenitor cell mobilization. Moreover, increased retention of the progenitor cells also resulted from up-regulated levels of stem cell factor (SCF) in the BM, in particular in the stem cell-rich endosteum and endothelial regions. Increased SCF-induced adhesion of primitive Sca-1(+)/c-Kit(+)/Lin(-) cells to osteoblasts was also the result of elevation of the receptor c-Kit. Regulation of these phenomena is mediated by hyperphosphorylation of c-Myc in hematopoietic progenitors of hpa-Tg mice or after exogenous heparanase addition to wildtype BM cells in vitro. Altogether, our data suggest that heparanase modification of the BM microenvironment regulates the retention and proliferation of hematopoietic progenitor cells.


Journal of Molecular Medicine | 2011

MT1-MMP and RECK: opposite and essential roles in hematopoietic stem and progenitor cell retention and migration

Karin Golan; Yaron Vagima; Polina Goichberg; Shiri Gur-Cohen; Tsvee Lapidot

Migratory capacity is a fundamental property of hematopoietic stem and progenitor cells (HSPCs). This feature is employed in clinical mobilization of HSPCs to the circulation and constitutes the basis for modern bone marrow (BM) transplantation procedures which are routinely used to treat hematological malignancies. Therefore, characterization of new players in the complex process of HSPC motility in steady-state conditions as well as during stress situations is a major challenge. We report that while the metalloproteinase membrane type 1-metalloprotease (MT1-MMP) has an essential role in human HSPC trafficking during granulocyte colony-stimulating factor (G-CSF)-induced mobilization, its inhibitor reversion-inducing cysteine-rich protein with Kazal motifs (RECK) and the adhesion molecule CD44 are required for HSPC retention to the BM in steady-state conditions. The nervous system via Wnt signaling along with HGF/c-Met signaling and the complement cascade play a major role in regulating MT1-MMP increased activity, CD44 cleavage, and RECK-reduced expression during G-CSF-induced mobilization. This review will elaborate on the opposite roles of MT1-MMP and RECK in HSPC migration and retention and suggest targeting them in order to facilitate HSPC mobilization and engraftment upon BM transplantation in patients.


Blood | 2010

The Chemotactic Lipid S1P Regulates Hematopoietic Progenitor Cell Egress and Mobilization Via Its Major Receptor S1P 1 and by SDF-1 Inhibition In a p38/Akt/mTOR Dependent Manner

Karin Golan; Yaron Vagima; Aya Ludin; Tomer Itkin; Alexander Kalinkovich; Shiri Cohen-Gur; Orit Kollet; Amir Schajnovitz; Shoham Shivtiel; Tsvee Lapidot


Archive | 2011

hematopoietic progenitor cells via ROS signaling induced mobilization of - Enhanced c-Met activity promotes G-CSF

Massimo Aglietta; Wanda Piacibello; Tsvee Lapidot Buss; Elias Shezen; Tomer Itkin; Orit Kollet; Isabelle Petit; Andreas Trumpp; James G. Christensen; Shoham Shivtiel; Alexander Kalinkovich; Luisa Caione; Loretta Gammaitoni; Elisa Laurenti; Karin Golan; Simona Corso; Silvia Giordano; Amir Schajnovitz; Yaron Vagima


Archive | 2010

cells via modulation of the bone marrow microenvironment - /Lin + /c-Kit + Heparanase regulates retention and proliferation of primitive Sca-1

Neta Netzer; Karin Golan; Itay Shafat; Neta Ilan; Arnon Nagler; Israel Vlodavsky; Tsvee Lapidot; Asaf Spiegel; Eyal Zcharia; Yaron Vagima; Tomer Itkin; Alexander Kalinkovich; Ayelet Dar; Orit Kollet

Collaboration


Dive into the Yaron Vagima's collaboration.

Top Co-Authors

Avatar

Alexander Kalinkovich

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Karin Golan

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Tsvee Lapidot

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Tomer Itkin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Orit Kollet

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Shoham Shivtiel

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Amir Schajnovitz

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayelet Dar

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Elias Shezen

Weizmann Institute of Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge