Network


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

Hotspot


Dive into the research topics where Asaf Spiegel is active.

Publication


Featured researches published by Asaf Spiegel.


Nature Medicine | 2006

Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells

Orit Kollet; Ayelet Dar; Shoham Shivtiel; Alexander Kalinkovich; Kfir Lapid; Yejezkel Sztainberg; Melania Tesio; Robert M Samstein; Polina Goichberg; Asaf Spiegel; Ari Elson; Tsvee Lapidot

Here we investigated the potential role of bone-resorbing osteoclasts in homeostasis and stress-induced mobilization of hematopoietic progenitors. Different stress situations induced activity of osteoclasts (OCLs) along the stem cell–rich endosteum region of bone, secretion of proteolytic enzymes and mobilization of progenitors. Specific stimulation of OCLs with RANKL recruited mainly immature progenitors to the circulation in a CXCR4- and MMP-9–dependent manner; however, RANKL did not induce mobilization in young female PTPε-knockout mice with defective OCL bone adhesion and resorption. Inhibition of OCLs with calcitonin reduced progenitor egress in homeostasis, G-CSF mobilization and stress situations. RANKL-stimulated bone-resorbing OCLs also reduced the stem cell niche components SDF-1, stem cell factor (SCF) and osteopontin along the endosteum, which was associated with progenitor mobilization. Finally, the major bone-resorbing proteinase, cathepsin K, also cleaved SDF-1 and SCF. Our findings indicate involvement of OCLs in selective progenitor recruitment as part of homeostasis and host defense, linking bone remodeling with regulation of hematopoiesis.


Journal of Clinical Investigation | 2003

HGF, SDF-1, and MMP-9 are involved in stress-induced human CD34 + stem cell recruitment to the liver

Orit Kollet; Shoham Shivtiel; Yuan–Qing Chen; Jenny Suriawinata; Swan N. Thung; Mariana D. Dabeva; Joy Kahn; Asaf Spiegel; Ayelet Dar; Sarit Samira; Polina Goichberg; Alexander Kalinkovich; Fernando Arenzana-Seisdedos; Arnon Nagler; Izhar Hardan; Michel Revel; David A. Shafritz; Tsvee Lapidot

Hematopoietic stem cells rarely contribute to hepatic regeneration, however, the mechanisms governing their homing to the liver, which is a crucial first step, are poorly understood. The chemokine stromal cell-derived factor-1 (SDF-1), which attracts human and murine progenitors, is expressed by liver bile duct epithelium. Neutralization of the SDF-1 receptor CXCR4 abolished homing and engraftment of the murine liver by human CD34+ hematopoietic progenitors, while local injection of human SDF-1 increased their homing. Engrafted human cells were localized in clusters surrounding the bile ducts, in close proximity to SDF-1-expressing epithelial cells, and differentiated into albumin-producing cells. Irradiation or inflammation increased SDF-1 levels and hepatic injury induced MMP-9 activity, leading to both increased CXCR4 expression and SDF-1-mediated recruitment of hematopoietic progenitors to the liver. Unexpectedly, HGF, which is increased following liver injury, promoted protrusion formation, CXCR4 upregulation, and SDF-1-mediated directional migration by human CD34+ progenitors, and synergized with stem cell factor. Thus, stress-induced signals, such as increased expression of SDF-1, MMP-9, and HGF, recruit human CD34+ progenitors with hematopoietic and/or hepatic-like potential to the liver of NOD/SCID mice. Our results suggest the potential of hematopoietic CD34+/CXCR4+cells to respond to stress signals from nonhematopoietic injured organs as an important mechanism for tissue targeting and repair.


Nature Immunology | 2007

Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34 + cells through Wnt signaling

Asaf Spiegel; Shoham Shivtiel; Alexander Kalinkovich; Aya Ludin; Neta Netzer; Polina Goichberg; Yaara Azaria; Igor B. Resnick; Izhar Hardan; Herzel Ben-Hur; Arnon Nagler; Menachem Rubinstein; Tsvee Lapidot

Catecholamines are important regulators of homeostasis, yet their functions in hematopoiesis are poorly understood. Here we report that immature human CD34+ cells dynamically expressed dopamine and β2-adrenergic receptors, with higher expression in the primitive CD34+CD38lo population. The myeloid cytokines G-CSF and GM-CSF upregulated neuronal receptor expression on immature CD34+ cells. Treatment with neurotransmitters increased the motility, proliferation and colony formation of human progenitor cells, correlating with increased polarity, expression of the metalloproteinase MT1-MMP and activity of the metalloproteinase MMP-2. Treatment with catecholamines enhanced human CD34+ cell engraftment of NOD-SCID mice through Wnt signaling activation and increased cell mobilization and bone marrow Sca-1+c-Kit+Lin− cell numbers. Our results identify new functions for neurotransmitters and myeloid cytokines in the direct regulation of human and mouse progenitor cell migration and development.


Journal of Leukocyte Biology | 2008

A crosstalk between intracellular CXCR7 and CXCR4 involved in rapid CXCL12‐triggered integrin activation but not in chemokine‐triggered motility of human T lymphocytes and CD34+ cells

Tanja Nicole Hartmann; Valentin Grabovsky; Ronit Pasvolsky; Ziv Shulman; Eike C. Buss; Asaf Spiegel; Arnon Nagler; Tsvee Lapidot; Marcus Thelen; Ronen Alon

The chemokine CXCL12 promotes migration of human leukocytes, hematopoietic progenitors, and tumor cells. The binding of CXCL12 to its receptor CXCR4 triggers Gi protein signals for motility and integrin activation in many cell types. CXCR7 is a second, recently identified receptor for CXCL12, but its role as an intrinsic G‐protein‐coupled receptor (GPCR) has been debated. We report that CXCR7 fails to support on its own any CXCL12‐triggered integrin activation or motility in human T lymphocytes or CD34+ progenitors. CXCR7 is also scarcely expressed on the surface of both cell types and concentrates right underneath the plasma membrane with partial colocalization in early endosomes. Nevertheless, various specific CXCR7 blockers get access to this pool and attenuate the ability of CXCR4 to properly rearrange by surface‐bound CXCL12, a critical step in the ability of the GPCR to trigger optimal CXCL12‐mediated stimulation of integrin activation in T lymphocytes as well as in CD34+ cells. In contrast, CXCL12‐triggered CXCR4 signaling to early targets, such as Akt as well as CXCR4‐mediated chemotaxis, is insensitive to identical CXCR7 blocking. Our findings suggest that although CXCR7 is not an intrinsic signaling receptor for CXCL12 on lymphocytes or CD34+ cells, its blocking can be useful for therapeutic interference with CXCR4‐mediated activation of integrins.


Cancer Discovery | 2016

Neutrophils Suppress Intraluminal NK Cell–Mediated Tumor Cell Clearance and Enhance Extravasation of Disseminated Carcinoma Cells

Asaf Spiegel; Mary W. Brooks; Houshyar S; Ferenc Reinhardt; Michele Ardolino; Fessler E; Michelle B. Chen; Krall Ja; J. DeCock; Ioannis K. Zervantonakis; Alexandre Iannello; Yoshiko Iwamoto; Cortez-Retamozo; Roger D. Kamm; Mikael J. Pittet; David H. Raulet; Robert A. Weinberg

UNLABELLED Immune cells promote the initial metastatic dissemination of carcinoma cells from primary tumors. In contrast to their well-studied functions in the initial stages of metastasis, the specific roles of immunocytes in facilitating progression through the critical later steps of the invasion-metastasis cascade remain poorly understood. Here, we define novel functions of neutrophils in promoting intraluminal survival and extravasation at sites of metastatic dissemination. We show that CD11b(+)/Ly6G(+) neutrophils enhance metastasis formation via two distinct mechanisms. First, neutrophils inhibit natural killer cell function, which leads to a significant increase in the intraluminal survival time of tumor cells. Thereafter, neutrophils operate to facilitate extravasation of tumor cells through the secretion of IL1β and matrix metalloproteinases. These results identify neutrophils as key regulators of intraluminal survival and extravasation through their cross-talk with host cells and disseminating carcinoma cells. SIGNIFICANCE This study provides important insights into the systemic contributions of neutrophils to cancer metastasis by identifying how neutrophils facilitate intermediate steps of the invasion-metastasis cascade. We demonstrate that neutrophils suppress natural killer cell activity and increase extravasation of tumor cells. Cancer Discov; 6(6); 630-49. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 561.


Brain Behavior and Immunity | 2009

Blood-forming stem cells are nervous: Direct and indirect regulation of immature human CD34 + cells by the nervous system

Alexander Kalinkovich; Asaf Spiegel; Shoham Shivtiel; Orit Kollet; Noela Jordaney; Wanda Piacibello; Tsvee Lapidot

The nervous system regulates immunity through hormonal and neuronal routes as part of host defense and repair mechanism. Here, we review the emerging evidence for regulation of human hematopoietic stem and progenitor cells (HSPC) by the nervous system both directly and indirectly via their bone marrow (BM) niche-supporting stromal cells. Functional expression of several neurotransmitter receptors was demonstrated on HSPC, mainly on the more primitive CD34(+)/CD38(-/low) fraction. The myeloid cytokines, G-CSF and GM-CSF, dynamically upregulate neuronal receptor expression on human HSPC. This is followed by an increased response to neurotransmitters, leading to enhanced proliferation and motility of human CD34(+) progenitors, repopulation of the murine BM and their egress to the circulation. Importantly, recent observations showed rapid mobilization of human HSPC to high SDF-1 expressing ischemic tissues of stroke individuals followed by neoangiogenesis, neurological and functional recovery. Along with decreased levels of circulating immature CD34(+) cells and SDF-1 blood levels found in patients with early-stage Alzheimers disease, these findings suggest a possible involvement of human HSPC in brain homeostasis and thus their potential clinical applications in neuropathology.


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.


Nature Cell Biology | 2018

IL-1β inflammatory response driven by primary breast cancer prevents metastasis-initiating cell colonization

Zafira Castaño; Beatriz P. San Juan; Asaf Spiegel; Ayush Pant; Molly J. DeCristo; Tyler Laszewski; Jessalyn M. Ubellacker; Susanne R. Janssen; Anushka Dongre; Ferenc Reinhardt; Ayana Henderson; Ana Garcia del Rio; Ann M. Gifford; Zachary T. Herbert; John N. Hutchinson; Robert A. Weinberg; Christine L. Chaffer; Sandra S. McAllister

Lack of insight into mechanisms governing breast cancer metastasis has precluded the development of curative therapies. Metastasis-initiating cancer cells (MICs) are uniquely equipped to establish metastases, causing recurrence and therapeutic resistance. Using various metastasis models, we discovered that certain primary tumours elicit a systemic inflammatory response involving interleukin-1β (IL-1β)-expressing innate immune cells that infiltrate distant MIC microenvironments. At the metastatic site, IL-1β maintains MICs in a ZEB1-positive differentiation state, preventing MICs from generating highly proliferative E-cadherin-positive progeny. Thus, when the inherent plasticity of MICs is impeded, overt metastases cannot be established. Ablation of the pro-inflammatory response or inhibition of the IL-1 receptor relieves the differentiation block and results in metastatic colonization. Among patients with lymph node-positive breast cancer, high primary tumour IL-1β expression is associated with better overall survival and distant metastasis-free survival. Our data reveal complex interactions that occur between primary tumours and disseminated MICs that could be exploited to improve patient survival.Castaño et al. show that primary breast tumours drive an IL-1β -mediated inflammatory response that inhibits cellular plasticity and metastatic colonization of metastasis-initiating cells.


Cancer Research | 2016

Abstract PR13: Understanding the systemic interactions between primary tumors and disseminated tumor initiating cells

Zafira Castaño; Christine L. Chaffer; Asaf Spiegel; Ayush Pant; Andrea L. Richardson; Ferenc Reinhardt; Timothy Marsh; Susanne R. Janssen; Ann M. Gifford; Robert A. Weinberg; Sandra S. McAllister

The causes for breast cancer recurrence in the form of metastatic disease and the reasons why less than 1% of disseminated tumor cells form metastases are unknown. A number of studies have demonstrated that the aggressive cancer cell population capable of driving metastasis feature properties of the epithelial-mesenchymal transition (EMT) and tumor initiation (TI). We previously reported novel mechanisms by which systemic and microenvironmental factors enrich tumors for EMT and TI genes and demonstrated that recurrence rates are not strictly due to tumor cell intrinsic properties (Castano et al., Cancer Discovery 2013). Using a preclinical model of breast cancer during the early phases of metastatic disease, when patients harbor disseminated tumor initiating cells (TICs) in the periphery at the time of their primary diagnosis (McAllister SS et al., Cell 2008), we recently made a surprising, and seemingly contrasting discovery. Specifically, we found that certain primary tumors can inhibit progression of disseminated TICs into overt tumors, maintaining them in the TI state and slowing their proliferation and differentiation to form tumor tissue mass. We established that the primary tumor induce mobilization of IL1beta-expressing monocytes that are recruited to sites where TICs reside. At those sites, signaling via the IL1 receptor maintains the EMT/TI state of the disseminated cells, thus promoting disease indolence. Importantly, surgical removal of the primary tumor enables TICs at the secondary sites to exit the EMT/TI state and to produce robustly growing tumors. Confirming our xenograft results, we established that stromal expression of IL1β in patient tumors is associated with poor outcome. Collectively, these data highlight the profound impact a primary tumor can exert on metastasizing cells - in this case, by altering the systemic environment to the detriment of secondary tumor growth. Moreover, our data highlight a central role for IL1β in modulating tumor cell plasticity and suggest it may provide a novel avenue for targeting recurrent disease. Citation Format: Zafira Castano, Christine L. Chaffer, Asaf Spiegel, Ayush Pant, Andrea L. Richardson, Ferenc Reinhardt, Timothy Marsh, Susanne Janssen, Ann M. Gifford, Robert A. Weinberg, Sandra S. McAllister. Understanding the systemic interactions between primary tumors and disseminated tumor initiating cells. [abstract]. In: Proceedings of the AACR Special Conference: Function of Tumor Microenvironment in Cancer Progression; 2016 Jan 7–10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2016;76(15 Suppl):Abstract nr PR13.


Blood | 2001

Rapid and efficient homing of human CD34+CD38−/lowCXCR4+stem and progenitor cells to the bone marrow and spleen of NOD/SCID and NOD/SCID/B2mnull mice

Orit Kollet; Asaf Spiegel; Amnon Peled; Isabelle Petit; Tamara Byk; Rami Hershkoviz; Esther Guetta; Gad Barkai; Arnon Nagler; Tsvee Lapidot

Collaboration


Dive into the Asaf Spiegel's collaboration.

Top Co-Authors

Avatar

Tsvee Lapidot

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Alexander Kalinkovich

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Arnon Nagler

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Shoham Shivtiel

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Menachem Rubinstein

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Amnon Peled

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Ayelet Dar

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Polina Goichberg

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Ferenc Reinhardt

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge