Network


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

Hotspot


Dive into the research topics where Aya Ludin is active.

Publication


Featured researches published by Aya Ludin.


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.


Nature | 2016

Distinct bone marrow blood vessels differentially regulate haematopoiesis.

Tomer Itkin; Shiri Gur-Cohen; Joel A. Spencer; Amir Schajnovitz; Saravana K. Ramasamy; Anjali P. Kusumbe; Guy Ledergor; Yookyung Jung; Idan Milo; Michael G. Poulos; Alexander Kalinkovich; Aya Ludin; Orit Kollet; Guy Shakhar; Jason M. Butler; Shahin Rafii; Ralf H. Adams; David T. Scadden; Charles P. Lin; Tsvee Lapidot

Bone marrow endothelial cells (BMECs) form a network of blood vessels that regulate both leukocyte trafficking and haematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance these dual roles, and whether these events occur at the same vascular site. We found that mammalian bone marrow stem cell maintenance and leukocyte trafficking are regulated by distinct blood vessel types with different permeability properties. Less permeable arterial blood vessels maintain haematopoietic stem cells in a low reactive oxygen species (ROS) state, whereas the more permeable sinusoids promote HSPC activation and are the exclusive site for immature and mature leukocyte trafficking to and from the bone marrow. A functional consequence of high permeability of blood vessels is that exposure to blood plasma increases bone marrow HSPC ROS levels, augmenting their migration and differentiation, while compromising their long-term repopulation and survival. These findings may have relevance for clinical haematopoietic stem cell transplantation and mobilization protocols.


Leukemia | 2011

Rapid mobilization of hematopoietic progenitors by AMD3100 and catecholamines is mediated by CXCR4-dependent SDF-1 release from bone marrow stromal cells

Ayelet Dar; Amir Schajnovitz; Kfir Lapid; Alexander Kalinkovich; Tomer Itkin; Aya Ludin; Wei-Ming Kao; Michela Battista; Melania Tesio; Orit Kollet; Neta Netzer Cohen; Raanan Margalit; Eike C. Buss; Françoise Baleux; Shinya Oishi; Nobutaka Fujii; Andre Larochelle; Cynthia E. Dunbar; Hal E. Broxmeyer; Paul S. Frenette; Tsvee Lapidot

Steady-state egress of hematopoietic progenitor cells can be rapidly amplified by mobilizing agents such as AMD3100, the mechanism, however, is poorly understood. We report that AMD3100 increased the homeostatic release of the chemokine stromal cell derived factor-1 (SDF-1) to the circulation in mice and non-human primates. Neutralizing antibodies against CXCR4 or SDF-1 inhibited both steady state and AMD3100-induced SDF-1 release and reduced egress of murine progenitor cells over mature leukocytes. Intra-bone injection of biotinylated SDF-1 also enhanced release of this chemokine and murine progenitor cell mobilization. AMD3100 directly induced SDF-1 release from CXCR4+ human bone marrow osteoblasts and endothelial cells and activated uPA in a CXCR4/JNK-dependent manner. Additionally, ROS inhibition reduced AMD3100-induced SDF-1 release, activation of circulating uPA and mobilization of progenitor cells. Norepinephrine treatment, mimicking acute stress, rapidly increased SDF-1 release and progenitor cell mobilization, whereas β2-adrenergic antagonist inhibited both steady state and AMD3100-induced SDF-1 release and progenitor cell mobilization in mice. In conclusion, this study reveals that SDF-1 release from bone marrow stromal cells to the circulation emerges as a pivotal mechanism essential for steady-state egress and rapid mobilization of hematopoietic progenitor cells, but not mature leukocytes.


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.


Nature Immunology | 2012

Monocytes-macrophages that express α-smooth muscle actin preserve primitive hematopoietic cells in the bone marrow

Aya Ludin; Tomer Itkin; Shiri Gur-Cohen; Alexander Mildner; Elias Shezen; Karin Golan; Orit Kollet; Alexander Kalinkovich; Ziv Porat; Gabriele D'Uva; Amir Schajnovitz; Elena Voronov; David A Brenner; Ron N. Apte; Steffen Jung; Tsvee Lapidot

Hematopoietic stem and progenitor cells (HSPCs) are regulated by various bone marrow stromal cell types. Here we identified rare activated bone marrow monocytes and macrophages with high expression of α-smooth muscle actin (α-SMA) and the cyclooxygenase COX-2 that were adjacent to primitive HSPCs. These myeloid cells resisted radiation-induced cell death and further upregulated COX-2 expression under stress conditions. COX-2-derived prostaglandin E2 (PGE2) prevented HSPC exhaustion by limiting the production of reactive oxygen species (ROS) via inhibition of the kinase Akt and higher stromal-cell expression of the chemokine CXCL12, which is essential for stem-cell quiescence. Our study identifies a previously unknown subset of α-SMA+ activated monocytes and macrophages that maintain HSPCs and protect them from exhaustion during alarm situations.


Nature Immunology | 2011

CXCL12 secretion by bone marrow stromal cells is dependent on cell contact and mediated by connexin-43 and connexin-45 gap junctions

Amir Schajnovitz; Tomer Itkin; Gabriele D'Uva; Alexander Kalinkovich; Karin Golan; Aya Ludin; Dror Cohen; Ziv Shulman; Abraham Avigdor; Arnon Nagler; Orit Kollet; Rony Seger; Tsvee Lapidot

The chemokine CXCL12 is essential for the function of hematopoietic stem and progenitor cells. Here we report that secretion of functional CXCL12 from human bone marrow stromal cells (BMSCs) was a cell contact–dependent event mediated by connexin-43 (Cx43) and Cx45 gap junctions. Inhibition of connexin gap junctions impaired the secretion of CXCL12 and homing of leukocytes to mouse bone marrow. Purified human CD34+ progenitor cells did not adhere to noncontacting BMSCs, which led to a much smaller pool of immature cells. Calcium conduction activated signaling by cAMP–protein kinase A (PKA) and induced CXCL12 secretion mediated by the GTPase RalA. Cx43 and Cx45 additionally controlled Cxcl12 transcription by regulating the nuclear localization of the transcription factor Sp1. We suggest that BMSCs form a dynamic syncytium via connexin gap junctions that regulates CXC12 secretion and the homeostasis of hematopoietic stem cells.


Antioxidants & Redox Signaling | 2014

Reactive Oxygen Species Regulate Hematopoietic Stem Cell Self-Renewal, Migration and Development, As Well As Their Bone Marrow Microenvironment

Aya Ludin; Shiri Gur-Cohen; Karin Golan; Kerstin B. Kaufmann; Tomer Itkin; Chiara Medaglia; Xin-Jiang Lu; Guy Ledergor; Orit Kollet; Tsvee Lapidot

SIGNIFICANCE Blood forming, hematopoietic stem cells (HSCs) mostly reside in the bone marrow in a quiescent, nonmotile state via adhesion interactions with stromal cells and macrophages. Quiescent, proliferating, and differentiating stem cells have different metabolism, and accordingly different amounts of intracellular reactive oxygen species (ROS). Importantly, ROS is not just a byproduct of metabolism, but also plays a role in stem cell state and function. RECENT ADVANCES ROS levels are dynamic and reversibly dictate enhanced cycling and myeloid bias in ROS(high) short-term repopulating stem cells, and ROS(low) quiescent long-term repopulating stem cells. Low levels of ROS, regulated by intrinsic factors such as cell respiration or nicotinamide adenine dinucleotide phosphate-oxidase (NADPH oxidase) activity, or extrinsic factors such as stem cell factor or prostaglandin E2 are required for maintaining stem cell self-renewal. High ROS levels, due to stress and inflammation, induce stem cell differentiation and enhanced motility. CRITICAL ISSUES Stem cells need to be protected from high ROS levels to avoid stem cell exhaustion, insufficient host immunity, and leukemic transformation that may occur during chronic inflammation. However, continuous low ROS production will lead to lack of stem cell function and opportunistic infections. Ultimately, balanced ROS levels are crucial for maintaining the small stem cell pool and host immunity, both in homeostasis and during stress situations. FUTURE DIRECTIONS Deciphering the signaling pathway of ROS in HSC will provide a better understanding of ROS roles in switching HSC from quiescence to activation and vice versa, and will also shed light on the possible roles of ROS in leukemia initiation and development.


Blood | 2012

FGF-2 expands murine hematopoietic stem and progenitor cells via proliferation of stromal cells, c-Kit activation, and CXCL12 down-regulation

Tomer Itkin; Aya Ludin; Ben Gradus; Shiri Gur-Cohen; Alexander Kalinkovich; Amir Schajnovitz; Orit Kollet; Jonathan Canaani; Elias Shezen; Douglas J. Coffin; Grigori Enikolopov; Thorsten Berg; Wanda Piacibello; Eran Hornstein; Tsvee Lapidot

Cytokine-induced expansion of hematopoietic stem and progenitor cells (HSPCs) is not fully understood. In the present study, we show that whereas steady-state hematopoiesis is normal in basic fibroblast growth factor (FGF-2)-knockout mice, parathyroid hormone stimulation and myeloablative treatments failed to induce normal HSPC proliferation and recovery. In vivo FGF-2 treatment expanded stromal cells, including perivascular Nestin(+) supportive stromal cells, which may facilitate HSPC expansion by increasing SCF and reducing CXCL12 via mir-31 up-regulation. FGF-2 predominantly expanded a heterogeneous population of undifferentiated HSPCs, preserving and increasing durable short- and long-term repopulation potential. Mechanistically, these effects were mediated by c-Kit receptor activation, STAT5 phosphorylation, and reduction of reactive oxygen species levels. Mice harboring defective c-Kit signaling exhibited abrogated HSPC expansion in response to FGF-2 treatment, which was accompanied by elevated reactive oxygen species levels. The results of the present study reveal a novel mechanism underlying FGF-2-mediated in vivo expansion of both HSPCs and their supportive stromal cells, which may be used to improve stem cell engraftment after clinical transplantation.


Nature Medicine | 2015

PAR1 signaling regulates the retention and recruitment of EPCR-expressing bone marrow hematopoietic stem cells

Shiri Gur-Cohen; Tomer Itkin; Sagarika Chakrabarty; Claudine Graf; Orit Kollet; Aya Ludin; Karin Golan; Alexander Kalinkovich; Guy Ledergor; Eitan Wong; Elisabeth Niemeyer; Ziv Porat; Ayelet Erez; Irit Sagi; Charles T. Esmon; Wolfram Ruf; Tsvee Lapidot

Retention of long-term repopulating hematopoietic stem cells (LT-HSCs) in the bone marrow is essential for hematopoiesis and for protection from myelotoxic injury. We report that signaling cascades that are traditionally viewed as coagulation related also control retention of endothelial protein C receptor–positive (EPCR+) LT-HSCs in the bone marrow and their recruitment to the blood via two pathways mediated by protease activated receptor 1 (PAR1). Thrombin-PAR1 signaling induces nitric oxide (NO) production, leading to EPCR shedding mediated by tumor necrosis factor-α–converting enzyme (TACE), enhanced CXCL12-CXCR4–induced motility and rapid stem and progenitor cell mobilization. Conversely, bone marrow blood vessels provide a microenvironment enriched with activated protein C (aPC) that retains EPCR+ LT-HSCs by limiting NO generation, reducing Cdc42 activity and enhancing integrin VLA4 affinity and adhesion. Inhibition of NO production by aPC-EPCR-PAR1 signaling reduces progenitor cell egress from the bone marrow, increases retention of bone marrow NOlow EPCR+ LT-HSCs and protects mice from chemotherapy-induced hematological failure and death. Our study reveals new roles for PAR1 and EPCR in controlling NO production to balance maintenance and recruitment of bone marrow EPCR+ LT-HSCs, with potential clinical relevance for stem cell transplantation.


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.

Collaboration


Dive into the Aya Ludin's collaboration.

Top Co-Authors

Avatar

Tomer Itkin

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

Tsvee Lapidot

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

Karin Golan

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Shiri Gur-Cohen

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

Kfir Lapid

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Guy Ledergor

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