68 results on '"Tomer Itkin"'
Search Results
2. Editorial: The Dynamic Interface Between Vascular Blood Vessels to Blood Forming Hematopoietic Stem Cells in Health and Disease
- Author
-
Tomer Itkin, Delfim Duarte, and Diana Passaro
- Subjects
hematopoeific stem cells ,vascular niche ,hematopoiesis ,endothelial cell (EC) ,malignant hematopoiesis ,developmental hematopoiesis ,Biology (General) ,QH301-705.5 - Published
- 2022
- Full Text
- View/download PDF
3. Lactate released by inflammatory bone marrow neutrophils induces their mobilization via endothelial GPR81 signaling
- Author
-
Eman Khatib-Massalha, Suditi Bhattacharya, Hassan Massalha, Adi Biram, Karin Golan, Orit Kollet, Anju Kumari, Francesca Avemaria, Ekaterina Petrovich-Kopitman, Shiri Gur-Cohen, Tomer Itkin, Isabell Brandenburger, Asaf Spiegel, Ziv Shulman, Zachary Gerhart-Hines, Shalev Itzkovitz, Matthias Gunzer, Stefan Offermanns, Ronen Alon, Amiram Ariel, and Tsvee Lapidot
- Subjects
Science - Abstract
Lactate is a by-product of glycolysis that can function via its G protein receptor GPR81. Here the authors show that LPS or Salmonella infection enhances glycolytic metabolism in bone marrow neutrophils, resulting in lactate production, which increases endothelial barrier permeability and mobilization of these neutrophils by targeting endothelial GPR81.
- Published
- 2020
- Full Text
- View/download PDF
4. Bioengineering the Bone Marrow Vascular Niche
- Author
-
Thomas Bessy, Tomer Itkin, and Diana Passaro
- Subjects
bone marrow ,vascular niche ,endothelial cells ,bioengineering ,bioprinting ,manufacturing ,Biology (General) ,QH301-705.5 - Abstract
The bone marrow (BM) tissue is the main physiological site for adult hematopoiesis. In recent years, the cellular and matrix components composing the BM have been defined with unprecedent resolution, both at the molecular and structural levels. With the expansion of this knowledge, the possibility of reproducing a BM-like structure, to ectopically support and study hematopoiesis, becomes a reality. A number of experimental systems have been implemented and have displayed the feasibility of bioengineering BM tissues, supported by cells of mesenchymal origin. Despite being known as an abundant component of the BM, the vasculature has been largely disregarded for its role in regulating tissue formation, organization and determination. Recent reports have highlighted the crucial role for vascular endothelial cells in shaping tissue development and supporting steady state, emergency and malignant hematopoiesis, both pre- and postnatally. Herein, we review the field of BM-tissue bioengineering with a particular focus on vascular system implementation and integration, starting from describing a variety of applicable in vitro models, ending up with in vivo preclinical models. Additionally, we highlight the challenges of the field and discuss the clinical perspectives in terms of adoptive transfer of vascularized BM-niche grafts in patients to support recovering hematopoiesis.
- Published
- 2021
- Full Text
- View/download PDF
5. Cooperative ETS transcription factors enforce adult endothelial cell fate and cardiovascular homeostasis
- Author
-
Jesus M. Gomez-Salinero, Tomer Itkin, Sean Houghton, Chaitanya Badwe, Yang Lin, Viktoria Kalna, Neil Dufton, Claire R. Peghaire, Masataka Yokoyama, Matthew Wingo, Tyler M. Lu, Ge Li, Jenny Zhaoying Xiang, Yen-Michael Sheng Hsu, David Redmond, Ryan Schreiner, Graeme M. Birdsey, Anna M. Randi, and Shahin Rafii
- Subjects
Article - Abstract
Current dogma dictates that, during adulthood, endothelial cells (ECs) are locked in an immutable stable homeostatic state. By contrast, herein we show that maintenance of EC fate and function are linked and active processes, which depend on the constitutive cooperativity of only two ETS transcription factors (TFs), ERG and Fli1. Although deletion of either ERG or Fli1 manifests subtle vascular dysfunction, their combined genetic deletion in adult ECs results in acute vasculopathy and multi-organ failure, due to loss of EC fate and integrity, hyperinflammation and spontaneous thrombosis, leading to death. ERG and Fli1 co-deficiency causes rapid transcriptional silencing of pan and organotypic vascular core genes, with dysregulation of inflammation and coagulation pathways. Vascular hyperinflammation leads to impaired hematopoiesis with myeloid skewing. Accordingly, enforced ERG and FLI1 expression in adult human mesenchymal stromal cells activates vascular programs and functionality, enabling in vivo engraftment of a perfusable vascular network. Genome-wide association study analysis identified vascular diseases that are associated with FLI1/ERG mutations. Constitutive expression of ERG and Fli1 upholds EC fate, physiological function and resilience in adult vasculature, whereas their functional loss can contribute to systemic human diseases.
- Published
- 2022
6. Single Cell Resolution Spatial Mapping of Human Hematopoiesis Reveals Aging-Associated Topographic Remodeling
- Author
-
Aleksandr Sarachakov, Arina Varlamova, Viktor Svekolkin, Ilia Galkin, Itzel Valencia, Caitlin Unkenholz, Tania Pannellini, Aida Akaeva, Sofia Smirnova, Pavel Ovcharov, Margarita Polyakova, Dmitrii Tabakov, Ekaterina Postovalova, Isha Sethi, Nara Shin, Alexander Bagaev, Tomer Itkin, Genevieve Crane, Michael Kluk, Julia Geyer, Giorgio Inghirami, and Sanjay Patel
- Abstract
The spatial anatomy of hematopoiesis in bone marrow has been extensively studied in mice and other preclinical models, but technical challenges have precluded a commensurate exploration in humans. Institutional pathology archives contain thousands of paraffinized bone marrow core biopsy tissue specimens, providing a rich resource for studying the intact human bone marrow topography in a variety of physiologic states. Thus, we developed an end-to-end pipeline involving multiparameter whole tissue staining, in situ imaging at single-cell resolution, and artificial intelligence (AI)-based digital image analysis, and then applied it to a cohort of disease-free samples to survey alterations in the hematopoietic topography associated with aging. Our data indicate heterogeneity in marrow adipose tissue (MAT) content within each age group, and an inverse correlation between MAT content and proportions of early myeloid and erythroid precursors, irrespective of age. We identify consistent endosteal and perivascular positioning of hematopoietic stem and progenitor cells (HSPCs) with medullary localization of more differentiated elements and, importantly, uncover new evidence of aging-associated changes in cellular and vascular morphologies, microarchitectural alterations suggestive of inflammaging, and diminution of a potentially active megakaryocytic niche. Overall, our findings suggest that there is topographic remodeling of human hematopoiesis associated with aging. More generally, we demonstrate the potential to deeply unravel the spatial biology of normal and pathologic human bone marrow states using intact archival tissue specimens.
- Published
- 2023
7. Cardiovascular diseases disrupt the bone-marrow niche
- Author
-
Tomer Itkin and Shahin Rafii
- Subjects
Multidisciplinary - Published
- 2021
8. Lactate released by inflammatory bone marrow neutrophils induces their mobilization via endothelial GPR81 signaling
- Author
-
Asaf Spiegel, Hassan Massalha, Shalev Itzkovitz, Orit Kollet, Eman Khatib-Massalha, Isabell Brandenburger, Suditi Bhattacharya, Francesca Avemaria, Ronen Alon, Ziv Shulman, Zachary Gerhart-Hines, Amiram Ariel, Tsvee Lapidot, Tomer Itkin, Adi Biram, Karin Golan, Ekaterina Petrovich-Kopitman, Anju Kumari, Stefan Offermanns, Matthias Gunzer, Shiri Gur-Cohen, Biram, Adi [0000-0001-6169-9861], Shulman, Ziv [0000-0002-9604-212X], Itzkovitz, Shalev [0000-0003-0685-2522], Gunzer, Matthias [0000-0002-5534-6055], Offermanns, Stefan [0000-0001-8676-6805], Ariel, Amiram [0000-0002-7469-5728], Lapidot, Tsvee [0000-0001-9844-6454], and Apollo - University of Cambridge Repository
- Subjects
Lipopolysaccharides ,Male ,Salmonella typhimurium ,0301 basic medicine ,Neutrophils ,Medizin ,General Physics and Astronomy ,HYPOXIA ,Vascular permeability ,GPR81 ,ANGIOGENESIS ,Receptors, G-Protein-Coupled ,ACTIVATION ,Mice ,0302 clinical medicine ,Bone Marrow ,NADPH OXIDASE ,TRANSCRIPTION ,Acute inflammation ,lcsh:Science ,Mice, Knockout ,chemistry.chemical_classification ,Multidisciplinary ,Cell biology ,CXCL1 ,CXCL2 ,medicine.anatomical_structure ,CHEMOKINES ,030220 oncology & carcinogenesis ,Salmonella Infections ,Female ,medicine.symptom ,Signal Transduction ,Endothelium ,Science ,Bone Marrow Cells ,Inflammation ,G-CSF ,METABOLISM ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Animals ,Humans ,Lactic Acid ,Reactive oxygen species ,CELL MOBILIZATION ,RECEPTOR ,General Chemistry ,Disease Models, Animal ,Metabolism ,030104 developmental biology ,chemistry ,lcsh:Q ,Endothelium, Vascular ,Bone marrow ,Bacterial infection - Abstract
Neutrophils provide first line of host defense against bacterial infections utilizing glycolysis for their effector functions. How glycolysis and its major byproduct lactate are triggered in bone marrow (BM) neutrophils and their contribution to neutrophil mobilization in acute inflammation is not clear. Here we report that bacterial lipopolysaccharides (LPS) or Salmonella Typhimurium triggers lactate release by increasing glycolysis, NADPH-oxidase-mediated reactive oxygen species and HIF-1α levels in BM neutrophils. Increased release of BM lactate preferentially promotes neutrophil mobilization by reducing endothelial VE-Cadherin expression, increasing BM vascular permeability via endothelial lactate-receptor GPR81 signaling. GPR81−/− mice mobilize reduced levels of neutrophils in response to LPS, unless rescued by VE-Cadherin disrupting antibodies. Lactate administration also induces release of the BM neutrophil mobilizers G-CSF, CXCL1 and CXCL2, indicating that this metabolite drives neutrophil mobilization via multiple pathways. Our study reveals a metabolic crosstalk between lactate-producing neutrophils and BM endothelium, which controls neutrophil mobilization under bacterial infection., Lactate is a by-product of glycolysis that can function via its G protein receptor GPR81. Here the authors show that LPS or Salmonella infection enhances glycolytic metabolism in bone marrow neutrophils, resulting in lactate production, which increases endothelial barrier permeability and mobilization of these neutrophils by targeting endothelial GPR81.
- Published
- 2020
9. Histone variant H3.3 maintains adult haematopoietic stem cell homeostasis by enforcing chromatin adaptability
- Author
-
Peipei Guo, Ying Liu, Fuqiang Geng, Andrew W. Daman, Xiaoyu Liu, Liangwen Zhong, Arjun Ravishankar, Raphael Lis, José Gabriel Barcia Durán, Tomer Itkin, Fanying Tang, Tuo Zhang, Jenny Xiang, Koji Shido, Bi-sen Ding, Duancheng Wen, Steven Z. Josefowicz, and Shahin Rafii
- Subjects
Mice, Knockout ,Myelopoiesis ,Macrophages ,Cell Cycle Proteins ,Cell Biology ,CD8-Positive T-Lymphocytes ,Hematopoietic Stem Cells ,Methylation ,Chromatin ,Article ,Cell Line ,Hematopoiesis ,Histones ,Mice, Inbred C57BL ,Mice ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Histone Chaperones ,Promoter Regions, Genetic ,Protein Processing, Post-Translational ,Granulocytes ,Transcription Factors - Abstract
Histone variants and the associated post-translational modifications that govern the stemness of haematopoietic stem cells (HSCs) and differentiation thereof into progenitors (HSPCs) have not been well defined. H3.3 is a replication-independent H3 histone variant in mammalian systems that is enriched at both H3K4me3- and H3K27me3-marked bivalent genes as well as H3K9me3-marked endogenous retroviral repeats. Here we show that H3.3, but not its chaperone Hira, prevents premature HSC exhaustion and differentiation into granulocyte-macrophage progenitors. H3.3-null HSPCs display reduced expression of stemness and lineage-specific genes with a predominant gain of H3K27me3 marks at their promoter regions. Concomitantly, loss of H3.3 leads to a reduction of H3K9me3 marks at endogenous retroviral repeats, opening up binding sites for the interferon regulatory factor family of transcription factors, allowing the survival of rare, persisting H3.3-null HSCs. We propose a model whereby H3.3 maintains adult HSC stemness by safeguarding the delicate interplay between H3K27me3 and H3K9me3 marks, enforcing chromatin adaptability.
- Published
- 2021
10. Publisher Correction: Histone variant H3.3 maintains adult haematopoietic stem cell homeostasis by enforcing chromatin adaptability
- Author
-
Peipei Guo, Ying Liu, Fuqiang Geng, Andrew W. Daman, Xiaoyu Liu, Liangwen Zhong, Arjun Ravishankar, Raphael Lis, José Gabriel Barcia Durán, Tomer Itkin, Fanying Tang, Tuo Zhang, Jenny Xiang, Koji Shido, Bi-sen Ding, Duancheng Wen, Steven Z. Josefowicz, and Shahin Rafii
- Subjects
Cell Biology - Published
- 2022
11. 3002 – CIRCADIAN CUES REPROGRAM METABOLIC PATHWAYS IN LONG-TERM REPOPULATING HEMATOPOIETIC STEM CELLS
- Author
-
Priyasmita Chakrabarti, Orit Kollet, Karin Golan, Lizeth-Alejandra Ordonez-Moreno, Montaser Haddad, Ekaterina Petrovich-Kopitman, Iwo Kucinski, Nicola Wilson, Sarah Kinston, Stephanie Xie, Tomer Itkin, John Dick, Bertie Göttgens, and Tsvee Lapidot
- Subjects
Cancer Research ,Genetics ,Cell Biology ,Hematology ,Molecular Biology - Published
- 2022
12. Distinct bone marrow blood vessels differentially regulate haematopoiesis
- Author
-
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, Karin Golan, Eman Khatib, Anju Kumari, Orit Kollet, Guy Shakhar, Jason M. Butler, Shahin Rafii, Ralf H. Adams, David T. Scadden, Charles P. Lin, and Tsvee Lapidot
- Subjects
Male ,0301 basic medicine ,Nestin ,Mice ,Plasma ,0302 clinical medicine ,Bone Marrow ,Cell Movement ,Leukocyte Trafficking ,Leukocytes ,Antigens, Ly ,Cell Self Renewal ,Multidisciplinary ,Hematopoietic Stem Cell Transplantation ,Bone Marrow Stem Cell ,hemic and immune systems ,Cell Differentiation ,Arteries ,Research Highlight ,Hematopoietic Stem Cell Mobilization ,3. Good health ,Cell biology ,Endothelial stem cell ,Haematopoiesis ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Stem cell ,Receptors, CXCR4 ,Cell Survival ,Bone Marrow Cells ,Biology ,Permeability ,03 medical and health sciences ,medicine ,Animals ,Progenitor cell ,Endothelial Cells ,Membrane Proteins ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Hematopoiesis ,Mice, Inbred C57BL ,030104 developmental biology ,Immunology ,Blood Vessels ,Bone marrow ,Pericytes ,Reactive Oxygen Species - Abstract
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.
- Published
- 2016
13. PAR1 signaling regulates the retention and recruitment of EPCR-expressing bone marrow hematopoietic stem cells
- Author
-
Ayelet Erez, Elisabeth Niemeyer, Sagarika Chakrabarty, Guy Ledergor, Ziv Porat, Orit Kollet, Irit Sagi, Wolfram Ruf, Claudine Graf, Shiri Gur-Cohen, Alexander Kalinkovich, Charles T. Esmon, Tsvee Lapidot, Eitan Wong, Tomer Itkin, Aya Ludin, and Karin Golan
- Subjects
Receptors, CXCR4 ,Receptors, Cell Surface ,ADAM17 Protein ,Integrin alpha4beta1 ,Biology ,Nitric Oxide ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Bone Marrow ,Cell Movement ,Cell Adhesion ,medicine ,Animals ,Receptor, PAR-1 ,Progenitor cell ,cdc42 GTP-Binding Protein ,Cell adhesion ,Endothelial protein C receptor ,Thrombin ,Endothelial Protein C Receptor ,General Medicine ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Cell biology ,Mice, Inbred C57BL ,Transplantation ,ADAM Proteins ,Haematopoiesis ,medicine.anatomical_structure ,Cdc42 GTP-Binding Protein ,Immunology ,Bone marrow ,Stem cell ,Protein C ,Signal Transduction - Abstract
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 NO(low) 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.
- Published
- 2015
14. Open the gates: vascular neurocrine signaling mobilizes hematopoietic stem and progenitor cells
- Author
-
Shahin Rafii, Jesus M. Gomez-Salinero, and Tomer Itkin
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Bone Marrow ,mental disorders ,medicine ,Neuropeptide Y ,Progenitor cell ,Neurotransmitter ,Hematopoietic Stem Cell Transplantation ,General Medicine ,Neuropeptide Y receptor ,Hematopoietic Stem Cells ,humanities ,Cell biology ,Transplantation ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Bone marrow ,Signal transduction ,Signal Transduction ,Research Article - Abstract
Endothelial cells (ECs) are components of the hematopoietic microenvironment and regulate hematopoietic stem and progenitor cell (HSPC) homeostasis. Cytokine treatments that cause HSPC trafficking to peripheral blood are associated with an increase in dipeptidylpeptidase 4/CD26 (DPP4/CD26), an enzyme that truncates the neurotransmitter neuropeptide Y (NPY). Here, we show that enzymatically altered NPY signaling in ECs caused reduced VE-cadherin and CD31 expression along EC junctions, resulting in increased vascular permeability and HSPC egress. Moreover, selective NPY2 and NPY5 receptor antagonists restored vascular integrity and limited HSPC mobilization, demonstrating that the enzymatically controlled vascular gateway specifically opens by cleavage of NPY by CD26 signaling via NPY2 and NPY5 receptors. Mice lacking CD26 or NPY exhibited impaired HSPC trafficking that was restored by treatment with truncated NPY. Thus, our results point to ECs as gatekeepers of HSPC trafficking and identify a CD26-mediated NPY axis that has potential as a pharmacologic target to regulate hematopoietic trafficking in homeostatic and stress conditions.
- Published
- 2017
15. Daily Onset of Light and Darkness Differentially Controls Hematopoietic Stem Cell Differentiation and Maintenance
- Author
-
Biana Bernshtein, Irit Sagi, Orit Kollet, Andrzej Ciechanowicz, John E. Dick, Eman Khatib-Massalha, Sylwia Rzeszotek, Eugenia Flores-Figueroa, Shiri Gur-Cohen, Francesca Avemaria, Tsvee Lapidot, Mayla Bertagna, Aya Ludin-Tal, Mohana Devi Subramaniam, Nathali Kaushansky, Regina P. Markus, Hassan Massalha, Tevie Mehlman, Mariusz Z. Ratajczak, Karin Golan, Simón Méndez-Ferrer, Anju Kumari, Tomer Itkin, Andrés García-García, Zulma S. Ferreira, Hui Cheng, Tomasz Janus, Stephanie Z. Xie, Ekaterina Petrovich-Kopitman, Alexander Brandis, Tao Cheng, Suditi Bhattacharya, Ferreira, Zulma S [0000-0001-6571-837X], Rzeszotek, Sylwia [0000-0002-2157-0315], Xie, Stephanie [0000-0002-0284-494X], Flores-Figueroa, Eugenia [0000-0002-7453-1125], Gur-Cohen, Shiri [0000-0002-6372-2284], Ciechanowicz, Andrzej K [0000-0003-0052-136X], Ratajczak, Mariusz Z [0000-0002-0071-0198], Méndez-Ferrer, Simón [0000-0002-9805-9988], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,bone marrow ,light and darkness ,Light ,stem cell repopulation potential ,hematopoietic stem and progenitor cells ,TNF ,Vascular permeability ,melatonin ,Biology ,norepinephrine ,Epigenesis, Genetic ,Melatonin ,Blood cell ,03 medical and health sciences ,Mice ,maintenance and retention ,Genetics ,medicine ,Animals ,Progenitor cell ,vascular permeability ,Cells, Cultured ,Hematopoietic stem cell differentiation ,differentiation and egress ,Cell Differentiation ,Cell Biology ,Darkness ,Hematopoietic Stem Cells ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Molecular Medicine ,Bone marrow ,Stem cell ,medicine.drug ,Transcription Factors - Abstract
Hematopoietic stem and progenitor cells (HSPCs) tightly couple maintenance of the bone marrow (BM) reservoir, including undifferentiated long-term repopulating hematopoietic stem cells (LT-HSCs), with intensive daily production of mature leukocytes and blood replenishment. We found two daily peaks of BM HSPC activity that are initiated by onset of light and darkness providing this coupling. Both peaks follow transient elevation of BM norepinephrine and TNF secretion, which temporarily increase HSPC reactive oxygen species (ROS) levels. Light-induced norepinephrine and TNF secretion augments HSPC differentiation and increases vascular permeability to replenish the blood. In contrast, darkness-induced TNF increases melatonin secretion to drive renewal of HSPCs and LT-HSC potential through modulating surface CD150 and c-Kit expression, increasing COX-2/αSMA+ macrophages, diminishing vascular permeability, and reducing HSPC ROS levels. These findings reveal that light- and darkness-induced daily bursts of norepinephrine, TNF, and melatonin within the BM are essential for synchronized mature blood cell production and HSPC pool repopulation.
- Published
- 2017
16. MicroRNA-155 promotes G-CSF-induced mobilization of murine hematopoietic stem and progenitor cells via propagation of CXCL12 signaling
- Author
-
Arefeh Rouhi, Florian Kuchenbauer, C Ludwig, Orit Kollet, Hartmut Geiger, Edith Schneider, Robert Brooks, Karin Golan, Tsvee Lapidot, William G. Kerr, Christopher M. Russo, Eran Hornstein, Eman Khatib-Massalha, Shiri Gur-Cohen, Tomer Itkin, Anju Kumari, and John D. Chisholm
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,Biology ,Polymorphism, Single Nucleotide ,miR-155 ,03 medical and health sciences ,Mice ,microRNA ,Granulocyte Colony-Stimulating Factor ,Animals ,Progenitor cell ,Hematopoietic Stem Cell Mobilization ,Mice, Knockout ,Hematology ,Hematopoietic Stem Cells ,biological factors ,Chemokine CXCL12 ,Cell biology ,Haematopoiesis ,MicroRNAs ,030104 developmental biology ,Oncology ,embryonic structures ,Immunology ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Stem cell ,Signal Transduction - Abstract
MicroRNA-155 promotes G-CSF-induced mobilization of murine hematopoietic stem and progenitor cells via propagation of CXCL12 signaling
- Published
- 2017
17. Reactive Oxygen Species Regulate Hematopoietic Stem Cell Self-Renewal, Migration and Development, As Well As Their Bone Marrow Microenvironment
- Author
-
Chiara Medaglia, Shiri Gur-Cohen, Kerstin B. Kaufmann, Tsvee Lapidot, Karin Golan, Orit Kollet, Tomer Itkin, Guy Ledergor, Aya Ludin, and Xin-Jiang Lu
- Subjects
Physiology ,Clinical Biochemistry ,Stem cell theory of aging ,Bone Marrow Cells ,Stem cell factor ,Biology ,Biochemistry ,Bone Marrow ,Cell Movement ,Cancer stem cell ,medicine ,Animals ,Humans ,Stem Cell Niche ,Molecular Biology ,Cell Proliferation ,General Environmental Science ,Inflammation ,Induced stem cells ,Cell Cycle ,Hematopoietic stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Hematopoietic Stem Cells ,Forum Review Articles ,Cell biology ,Endothelial stem cell ,medicine.anatomical_structure ,Hematologic Neoplasms ,General Earth and Planetary Sciences ,Stem cell ,Reactive Oxygen Species ,Adult stem cell - Abstract
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 ROShigh short-term repopulating stem cells, and ROSlow 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. Antioxid. Redox Signal. 21, 1605–1619.
- Published
- 2014
18. Fli-1 Transcriptionally Integrates Microenvironmental Cues Sensing By Self-Renewing Hematopoietic Stem and Progenitor Cells
- Author
-
Ying Liu, Yang Lin, Jae-Hung Shieh, Chaitanya R. Badwe, Shahin Rafii, Sean Houghton, Fuqiang Geng, Jesus M. Gomez-Salinero, Peipei Guo, Tomer Itkin, Yen-Michael S. Hsu, David Redmond, and Brandon Hadland
- Subjects
0301 basic medicine ,fungi ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Embryonic stem cell ,Cell biology ,Transplantation ,03 medical and health sciences ,Haematopoiesis ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,RUNX1 ,chemistry ,medicine ,Bone marrow ,Progenitor cell ,Stem cell ,Reprogramming ,030215 immunology - Abstract
During adulthood and embryogenesis, fate decisions of hematopoietic stem and progenitor cells (HSPCs), such as specification, self-renewal, and differentiation are tightly regulated by their neighboring niche cells. Moreover, distinct types of niches supply differential cues to direct alternative cell fates for HSPCs. Yet, currently the intrinsic mechanisms balancing HSPC response obliqueness to microenvironmental signals are unknown. Friend Leukemia integration-1 (Fli-1), is an ETS transcription factor expressed by vascular beds and hematopoietic lineages. Fli-1 belongs to the "heptad factors" which are hypothesized to specify and sustain a hematopoietic cell fate. While Fli-1 overexpression is linked to leukemia, the functional role Fli-1 plays in HSPC specification and maintenance remains undefined. We show that inducible deletion of Fli-1 using a Rosa-CreERT2 transgenic adult mice (Fli-1ROSAΔ), results in a rapid thrombocytopenia-associated mortality. Transplantation of Fli-1ROSAΔ bone marrow (BM) cells into WT recipients, to exclude vascular-mediated defects, followed by induction of Fli-1 deletion, resulted with the same phenotype. In a set of modulated competitive transplantation experiments (differential induction time points pre- or post-transplant), we observed defective ability of Fli-1ROSAΔ HSPCs to lodge, engraft, and to sustain hematopoiesis post repopulation. Fli-1 deficient HSPCs exhibited reduced quiescent cell cycling status, a hallmark of stemness, and displayed enhanced apoptosis. Thus, Fli-1 is essential for previously unrecognized cell-autonomous HSPC functions. To determine whether Fli-1 modulates HSPC specification, Fli-1 was conditionally deleted using a developmental VE-cadherin (CDH5)-Cre transgenic model (Fli-1CDH5Δ). This resulted with premature mortality of Fli-1CDH5Δ embryos, accompanied with a hemorrhagic phenotype. Reduced numbers of hematopoietic cells were still detected in the AGM of e10.5 Fli-1CDH5Δ embryos. Conditional Fli-1 deletion using a developmental hematopoietic Vav-1 Cre transgenic model (Fli-1Vav-1Δ) resulted again with premature mortality. Reduced presence of embryonic Fli-1Vav-1Δ liver HSPCs was observed at e12.5. We also applied two in vitro co-culture systems, to study Fli-1 in endothelial to hematopoietic transition (EHT). First, isolated hemogenic endothelial cells (HEC) from WT and Fli-1ROSAΔ embryos were co-cultured with AGM-derived vascular niche. HECs isolated from Fli-1ROSAΔ AGM were still able to convert to CD45+ cells, however these cells did not expand on a vascular niche. Secondly, we have applied an endothelial to hematopoietic reprogramming system in which isolated lung ECs are virally introduced with DOX inducible FosB, Gfi1, Runx1, and Spi1 (FGRS) factors and co-cultured with vascular niche cells. Both WT and Fli-1ROSAΔ ECs were able to acquire a hemogenic like state resulting with a final capacity to convert into hematopoietic cells. Again, Fli-1ROSAΔ cells displayed lesser numbers of CD45+ cells at the end point, presumably due to impaired interaction with the vascular niche. Indeed, reduced expansion capacity was observed both for mature CD45+ and for HSPC derived from Fli-1CDH5Δ AGM region. Adult Fli-1ROSAΔ HSPCs exhibited the same niche-dependent expansion defect. Induction of Fli-1 deletion in vitro in adult HSPCs revealed loss of dependency on vascular niche inductive signals, as no additive expansion effect was observed for Fli-1ROSAΔ HSPCs in the presence of a vascular niche. Hence, Fli-1 is essential for HSPC expansion rather than hematopoietic specification. Differential RNA-seq analysis combined with epigenetic studies of expanding WT and Fli-1ROSAΔ HSPCs, revealed dysregulation of Fli-1-controlled pathways involved in transduction of microenvironmental signals for self-renewal. Unexpectedly, H3K27Ac analysis, a marker for transcriptional priming, revealed increased global acetylation of Fli-1ROSAΔ HSPCs' chromatin. Therefor, Fli-1 may not only perform as transcription activator, but foremostly as a genomic suppressor via modulation of histone acetylation status. Decrypting the mechanism(s) by which Fli-1 orchestrates HSPC self-renewal, may promote an improved expansion protocol of human HSPC pre-transplantation, and provide additional insights for microenvironmental sensing by Fli-1-dependent leukemic cells. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
19. Acute Inflammation Induces Lactate Release By Bone Marrow Neutrophils That Promotes Their Mobilization Via Endothelial GPR81 Signaling
- Author
-
Amiram Ariel, Hassan Massalha, Francesca Avemaria, Ronen Alon, Shiri Gur-Cohen, Karin Golan, Orit Kollet, Abraham Avigdor, Eman Khatib-Massalha, Anju Kumari, Stefan Offermanns, Tsvee Lapidot, Tomer Itkin, Shalev Itzkovitz, and Suditi Bhattacharya
- Subjects
NADPH oxidase ,Innate immune system ,biology ,Endothelium ,Chemistry ,Monocyte ,Immunology ,Vascular permeability ,Inflammation ,Cell Biology ,Hematology ,GPR81 ,Granulocyte ,Biochemistry ,Molecular biology ,medicine.anatomical_structure ,biology.protein ,medicine ,medicine.symptom - Abstract
Innate immune neutrophils provide the first line of host defense against bacterial infections. Neutrophils under steady state rely almost entirely on glycolysis and exhibit very low levels of oxidative phosphorylation. The metabolite lactate has long been considered a "waste byproduct" of cell metabolism which accumulates during inflammation and sepsis. Increased plasma lactate levels in human patients is used as a marker for sepsis diagnosis. However, the direct effector actions of lactate, particularly in regulating neutrophil mobilization and function during inflammation has remained obscure. To better understand the metabolic consequences of BM neutrophil activation during the onset of inflammation, we tested how bacterial lipopolysaccharides (mimicking gram negative bacterial inflammation) introduced intraperitoneally (i.p.) affect neutrophil metabolism and mobilization. RNAseq of sorted BM neutrophils revealed that LPS-activated neutrophils upregulate enzymes catalyzing the first part of glycolysis (hexokinase and PFKL) and downregulate the expression of TCA cycle enzymatic genes. In addition, LPS enhanced neutrophil lactate production and release as indicated by higher levels of BM lactate and higher expression of LDHA and MCT4. In addition, LPS increased NADPH oxidase (NOX)-mediated reactive oxygen species and HIF-1α levels in BM neutrophils, which are up-stream of glycolytic enzymes and lactate production and release. Recently, we reported that i.p. lactate administration rapidly activated and mobilized neutrophils from BM to the circulation (ASH, 2017). To test if lactate acts preferentially on neutrophils, we also examined other types of hematopoietic cells. Interestingly, we found that lactate specifically and rapidly (i.e., within 4 hrs) mobilized neutrophils to the circulation whereas the levels of peripheral blood (PB) monocytes, lymphocytes, granulocyte monocyte progenitors (GMPs) and hematopoietic progenitor stem cells (LSK) were reduced following lactate administration. LPS treatment failed to mobilize activated ROShigh neutrophils to the PB in NOX-/- mice, while lactate administration partially rescued this defect following LPS treatment. Our data also reveal that the NOX/ROS axis operates upstream of lactate production in BM neutrophils since abnormal metabolic rates were found in NOX-/- neutrophils during the onset of the acute inflammatory responses. Moreover, we found that BM endothelial cells (BMEC) abundantly express the highly selective lactate receptor GPR81, and that neutrophil-released lactate increased BM vascular permeability via BMEC GPR81 signaling (ASH, 2017). Consistent with a role of the lactate/GPR81 axis in enhanced vascular permeability, we find that i.p. injected LPS reduced VE-Cadherin expression on highly permeable sBMECs in GPR81 dependent manner. Notably, neutralizing VE-Cadherin in GPR81-/- mice can rescue and elevate PB neutrophil levels, similarly to wild-type (WT) mice, suggesting that VE-Cadherin is downstream of GPR81 signaling and plays a role in neutrophil mobilization. Finally, to examine the potential clinical relevance of our findings, we infected WT, NOX-/- and GPR81-/- mice with Salmonella Typhimurium and found out that this pathogen drove high generation of ROS, elevated HIF-1αlevels, and triggered lactate production and release in WT BM neutrophils. In contrast, BM neutrophils of infected NOX-/- mice exhibited significantly lower HIF-1αand impaired lactate production and release. Consequently, WT mice infected with Salmonella had a higher levels of neutrophils in the blood, as compared to their NOX-/- or GPR81-/- mice counterparts. Altogether, our data reveal that the same regulatory mechanisms by which neutrophils respond to LPS challenges are used during bacterial infection with Salmonella. Our study highlights lactate released by BM neutrophils as a key pro-inflammatory stimulus of a novel immune-metabolic crosstalk which is triggered by infection and locally opens the BM vascular barrier to facilitate neutrophil mobilization and recruitment to sites of inflammation. Targeting this immune-metabolic crosstalk between lactate-producing neutrophils and the BM endothelium could be useful for the control of pathological neutrophil activation and mobilization during bacterial infections and help treatments of neutrophil related immune disorders. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
20. Physiologic corticosterone oscillations regulate murine hematopoietic stem/progenitor cell proliferation and CXCL12 expression by bone marrow stromal progenitors
- Author
-
Gabriele D'Uva, Karin Golan, Orit Kollet, Giulia Caglio, Adi Neufeld-Cohen, Aya Ludin, Shiri Gur-Cohen, Alexander Kalinkovich, Tsvee Lapidot, Kfir Lapid, Chiara Medaglia, Alon Chen, Elias Shezen, Diana Varol, Jonathan Canaani, Yaron Vagima, Tomer Itkin, Kollet O., Vagima Y., D'Uva G., Golan K., Canaani J., Itkin T., Gur-Cohen S., Kalinkovich A., Caglio G., Medaglia C., Ludin A., Lapid K., Shezen E., Neufeld-Cohen A., Varol D., Chen A., and Lapidot T.
- Subjects
endocrine system ,Cancer Research ,medicine.medical_specialty ,Chemokine ,Stromal cell ,Blotting, Western ,Biology ,Real-Time Polymerase Chain Reaction ,Receptors, Corticotropin-Releasing Hormone ,Mice ,Downregulation and upregulation ,Bone Marrow ,Cell Movement ,Internal medicine ,polycyclic compounds ,medicine ,Animals ,Hematopoiesi ,RNA, Messenger ,Progenitor cell ,CXCL12/CXCR4 ,Receptor ,Cells, Cultured ,Cell Proliferation ,Mice, Knockout ,Animal ,Reverse Transcriptase Polymerase Chain Reaction ,Hematopoietic Stem Cell ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Hematopoiesis ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,Endocrinology ,Oncology ,HSPC ,biology.protein ,Bone marrow ,Stromal Cells ,Signal transduction ,Corticosterone ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
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. © 2013 Macmillan Publishers Limited All rights reserved.
- Published
- 2013
21. Corrigendum: Age-dependent modulation of vascular niches for haematopoietic stem cells
- Author
-
Anjali P, Kusumbe, Saravana K, Ramasamy, Tomer, Itkin, Maarja Andaloussi, Mäe, Urs H, Langen, Christer, Betsholtz, Tsvee, Lapidot, and Ralf H, Adams
- Subjects
Article - Abstract
Blood vessels define local microenvironments in the skeletal system, play crucial roles in osteogenesis and provide niches for haematopoietic stem cells1–6. The properties of niche-forming vessels and their changes in the ageing organism remain incompletely understood. Here, we show that Notch signalling in endothelial cells leads to the expansion of haematopoietic stem cell niches in bone, which involves increases in CD31-positive capillaries and PDGFRβ-positive perivascular cells, arteriole formation, and elevation of cellular stem cell factor levels. While endothelial hypoxia-inducible factor signalling promotes some of these aspects, it fails to enhance vascular niche function because of lacking arterialization and expansion of PDGFRβ-positive cells. In ageing mice, niche-forming vessels in the skeletal system are strongly reduced but can be restored by activation of endothelial Notch signalling. These findings argue that vascular niches for haematopoietic stem cells are part of complex, age-dependent microenvironments involving multiple cell populations and vessel subtypes.
- Published
- 2016
22. Regulation of Hematopoiesis and Osteogenesis by Blood Vessel-Derived Signals
- Author
-
Saravana K. Ramasamy, Ralf H. Adams, Tomer Itkin, Anjali P. Kusumbe, Tsvee Lapidot, and Shiri Gur-Cohen
- Subjects
0301 basic medicine ,Stem cell factor ,Organ development ,Biology ,03 medical and health sciences ,Bone Marrow ,Osteogenesis ,medicine ,Animals ,Humans ,Regeneration (biology) ,Endothelial Cells ,Cell Biology ,Cell biology ,Hematopoiesis ,Endothelial stem cell ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Blood Vessels ,Bone marrow ,Stem cell ,Developmental Biology ,Blood vessel ,Signal Transduction - Abstract
In addition to their conventional role as a versatile transport system, blood vessels provide signals controlling organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular osteoprogenitor cells and thereby control bone formation. Blood vessels are also a critical component of niche microenvironments for hematopoietic stem cells. Here we discuss key pathways and factors controlling endothelial cell behavior in bone, the role of vessels in osteogenesis, and the nature of vascular stem cell niches in bone marrow.
- Published
- 2016
23. Age-dependent changes in the bone marrow microenvironment
- Author
-
Tsvee Lapidot, Anjali P. Kusumbe, Tomer Itkin, Ralf H. Adams, and Saravana K. Ramasamy
- Subjects
Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,medicine ,Age dependent ,General Medicine ,Bone marrow ,business - Published
- 2016
24. Age-dependent changes in the bone marrow microenvironment
- Author
-
Anjali Kusumbe, Saravana Ramasamy, Tomer Itkin, Tsvee Lapidot, and Ralf Adams
- Subjects
General Medicine - Published
- 2016
25. FGF signaling facilitates postinjury recovery of mouse hematopoietic system
- Author
-
Xi C. He, Chu-Xia Deng, Tomer Itkin, Tsvee Lapidot, John M. Perry, Jason T. Ross, Linheng Li, Aparna Venkatraman, Mark Hembree, Meng Zhao, and Jeffrey S. Haug
- Subjects
Male ,Antimetabolites, Antineoplastic ,Receptors, CXCR4 ,Hematopoiesis and Stem Cells ,Hematopoietic System ,Immunology ,Gene Expression ,Bone Marrow Cells ,Mice, Transgenic ,Biology ,Fibroblast growth factor ,Biochemistry ,CXCR4 ,Mice ,Cell Movement ,Conditional gene knockout ,Animals ,Receptor, Fibroblast Growth Factor, Type 1 ,Progenitor cell ,Cells, Cultured ,Cell Proliferation ,Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Fibroblast growth factor receptor 1 ,NF-kappa B ,Cell Biology ,Hematology ,Flow Cytometry ,Immunohistochemistry ,Molecular biology ,Cell biology ,Fibroblast Growth Factors ,Mice, Inbred C57BL ,Transplantation ,Haematopoiesis ,Female ,Fluorouracil ,Signal transduction ,Megakaryocytes ,Signal Transduction - Abstract
Previous studies have shown that fibroblast growth factor (FGF) signaling promotes hematopoietic stem and progenitor cell (HSPC) expansion in vitro. However, it is unknown whether FGF promotes HSPC expansion in vivo. Here we examined FGF receptor 1 (FGFR1) expression and investigated its in vivo function in HSPCs. Conditional knockout (CKO) of Fgfr1 did not affect phenotypical number of HSPCs and homeostatic hematopoiesis, but led to a reduced engraftment only in the secondary transplantation. When treated with 5-fluorouracil (5FU), the Fgfr1 CKO mice showed defects in both proliferation and subsequent mobilization of HSPCs. We identified megakaryocytes (Mks) as a major resource for FGF production, and further discovered a novel mechanism by which Mks underwent FGF-FGFR signaling dependent expansion to accelerate rapid FGF production under stress. Within HSPCs, we observed an up-regulation of nuclear factor κB and CXCR4, a receptor for the chemoattractant SDF-1, in response to bone marrow damage only in control but not in Fgfr1 CKO model, accounting for the corresponding defects in proliferation and migration of HSPCs. This study provides the first in vivo evidence that FGF signaling facilitates postinjury recovery of the mouse hematopoietic system by promoting proliferation and facilitating mobilization of HSPCs.
- Published
- 2012
26. FGF-2 expands murine hematopoietic stem and progenitor cells via proliferation of stromal cells, c-Kit activation, and CXCL12 down-regulation
- Author
-
Orit Kollet, Jonathan Canaani, Eran Hornstein, Elias Shezen, Aya Ludin, Thorsten Berg, Ben Gradus, Wanda Piacibello, Yossi Ovadya, Amir Schajnovitz, Grigori Enikolopov, Shiri Gur-Cohen, Alexander Kalinkovich, Tsvee Lapidot, Tomer Itkin, and Douglas J. Coffin
- Subjects
Stromal cell ,Immunology ,Basic fibroblast growth factor ,Down-Regulation ,Gene Expression ,Mice, Transgenic ,Biology ,Fibroblast growth factor ,Models, Biological ,Biochemistry ,Mice ,chemistry.chemical_compound ,STAT5 Transcription Factor ,Animals ,Phosphorylation ,Progenitor cell ,Cells, Cultured ,Bone Marrow Transplantation ,Cell Proliferation ,Mice, Knockout ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,Cell Biology ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Cell biology ,Mice, Inbred C57BL ,Transplantation ,Proto-Oncogene Proteins c-kit ,Haematopoiesis ,chemistry ,Parathyroid Hormone ,Fibroblast Growth Factor 2 ,Stromal Cells ,Stem cell ,Reactive Oxygen Species - Abstract
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.
- Published
- 2012
27. CXCL12 secretion by bone marrow stromal cells is dependent on cell contact and mediated by connexin-43 and connexin-45 gap junctions
- Author
-
Tomer Itkin, Orit Kollet, Tsvee Lapidot, Gabriele D'Uva, Arnon Nagler, Dror Cohen, Alexander Kalinkovich, Rony Seger, Ziv Shulman, Amir Schajnovitz, Aya Ludin, Karin Golan, Abraham Avigdor, Schajnovitz A., Itkin T., D'Uva G., Kalinkovich A., Golan K., Ludin A., Cohen D., Shulman Z., Avigdor A., Nagler A., Kollet O., Seger R., and Lapidot T.
- Subjects
Stromal cell ,Immunology ,CD34 ,Connexin ,Bone Marrow Cells ,Biology ,Connexins ,Mice ,Cell Movement ,Animals ,Humans ,Immunology and Allergy ,Progenitor cell ,Coculture Technique ,Mice, Knockout ,Animal ,Stromal Cell ,Gap Junctions ,Hematopoietic Stem Cell ,Mesenchymal Stem Cells ,Hematopoietic Stem Cells ,Cyclic AMP-Dependent Protein Kinases ,Immunohistochemistry ,Chemokine CXCL12 ,Coculture Techniques ,biological factors ,Mice, Inbred C57BL ,Endothelial stem cell ,Mesenchymal Stem Cell ,Microscopy, Fluorescence ,Cancer research ,Bone Marrow Cell ,Cyclic AMP-Dependent Protein Kinase ,Calcium ,ral GTP-Binding Proteins ,Gap Junction ,sense organs ,Stromal Cells ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Human ,Homing (hematopoietic) ,Adult stem cell - Abstract
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. © 2011 Nature America, Inc. All rights reserved.
- Published
- 2011
28. Heparanase regulates retention and proliferation of primitive Sca-1+/c-Kit+/Lin− cells via modulation of the bone marrow microenvironment
- Author
-
Alexander Kalinkovich, Orit Kollet, Arnon Nagler, Eyal Zcharia, Ayelet Dar, Tsvee Lapidot, Yaron Vagima, Karin Golan, Itay Shafat, Neta Ilan, Tomer Itkin, Israel Vlodavsky, Neta Netzer, and Asaf Spiegel
- Subjects
Chemokine ,Hematopoiesis and Stem Cells ,Immunology ,Bone Marrow Cells ,Mice, Transgenic ,Stem cell factor ,Biochemistry ,Immunophenotyping ,Mice ,Bone Marrow ,Cell Movement ,Cell Adhesion ,medicine ,Animals ,Heparanase ,Progenitor cell ,Cell adhesion ,Cell Proliferation ,Glucuronidase ,biology ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Neoplasm Proteins ,Haematopoiesis ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Bone marrow ,Stem cell ,Peptide Hydrolases - Abstract
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.
- Published
- 2008
29. Synaptojanin 2 is a druggable mediator of metastasis and the gene is overexpressed and amplified in breast cancer
- Author
-
Ronen Alon, Hava Gil-Henn, Rotem Ben-Hamo, Marcelo Ehrlich, Nir Ben-Chetrit, Yosef Yarden, Tomer Itkin, Wolfgang J. Koestler, Dalia Seger, Silvia Carvalho, Carlos Caldas, Maicol Mancini, Ali Abdul-Hai, Daniela Aleida Ferraro, David Chetrit, Fresia Pareja, Mattia Lauriola, Marc Symons, Kirti Shukla, Ziv Shulman, Tsvee Lapidot, Hadas Cohen-Dvashi, Cindy Körner, Fernanda Milanezi, Moshit Lindzen, Merav Kedmi, Fernando Schmitt, Stefan Wiemann, Haim Barr, Sol Efroni, Roslin Russell, Ben-Chetrit, Nir, Chetrit, David, Russell, Roslin, K('o)rner, Cindy, Mancini, Maicol, Abdul-Hai, Ali, Itkin, Tomer, Carvalho, Silvia, Cohen-Dvashi, Hada, Koestler, Wolfgang J., Shukla, Kirti, Lindzen, Moshit, Kedmi, Merav, Lauriola, Mattia, Shulman, Ziv, Barr, Haim, Seger, Dalia, Ferraro, Daniela A., Pareja, Fresia, Gil-Henn, Hava, Lapidot, Tsvee, Alon, Ronen, Milanezi, Fernanda, Symons, Marc, Ben-Hamo, Rotem, Efroni, Sol, Schmitt, Fernando, Wiemann, Stefan, Caldas, Carlo, Ehrlich, Marcelo, and Yarden, Yosef
- Subjects
Immunoblotting ,Gene Dosage ,Endocytic recycling ,Fluorescent Antibody Technique ,Breast Neoplasms ,Synaptojanin ,Mice, SCID ,Biochemistry ,Statistics, Nonparametric ,Metastasis ,Mice ,Breast cancer ,Growth factor receptor ,BREAST CANCER ,Cell Movement ,Cell Line, Tumor ,Drug Discovery ,medicine ,Image Processing, Computer-Assisted ,Animals ,Humans ,Epidermal growth factor receptor ,EGFR pathway ,Pseudopodia ,Neoplasm Metastasis ,RNA, Small Interfering ,Molecular Biology ,biology ,Cell migration ,Cell Biology ,medicine.disease ,Immunohistochemistry ,Phosphoric Monoester Hydrolases ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Immunology ,Invadopodia ,METASTASIS ,Podosomes ,Cancer research ,biology.protein ,Microscopy, Electron, Scanning ,Female - Abstract
Amplified HER2, which encodes a member of the epidermal growth factor receptor (EGFR) family, is a target of effective therapies against breast cancer. In search for similarly targetable genomic aberrations, we identified copy number gains in SYNJ2, which encodes the 5'-inositol lipid phosphatase synaptojanin 2, as well as overexpression in a small fraction of human breast tumors. Copy gain and overexpression correlated with shorter patient survival and a low abundance of the tumor suppressor microRNA miR-31. SYNJ2 promoted cell migration and invasion in culture and lung metastasis of breast tumor xenografts in mice. Knocking down SYNJ2 impaired the endocytic recycling of EGFR and the formation of cellular lamellipodia and invadopodia. Screening compound libraries identified SYNJ2-specific inhibitors that prevented cell migration but did not affect the related neural protein SYNJ1, suggesting that SYNJ2 is a potentially druggable target to block cancer cell migration.
- Published
- 2015
30. Age-dependent modulation of vascular niches for haematopoietic stem cells
- Author
-
Ralf H. Adams, Urs H. Langen, Anjali P. Kusumbe, Saravana K. Ramasamy, Tsvee Lapidot, Christer Betsholtz, Maarja Andaloussi Mäe, and Tomer Itkin
- Subjects
0301 basic medicine ,Male ,Aging ,Angiogenesis ,Notch signaling pathway ,Stem cell factor ,Cell Count ,Biology ,Bone and Bones ,Receptor, Platelet-Derived Growth Factor beta ,03 medical and health sciences ,Mice ,Growth factor receptor ,Arteriole ,Osteogenesis ,medicine.artery ,medicine ,Animals ,Stem Cell Niche ,Stem Cell Factor ,Multidisciplinary ,Receptors, Notch ,Endothelial Cells ,Hematopoietic Stem Cells ,Cell biology ,Capillaries ,Endothelial stem cell ,Platelet Endothelial Cell Adhesion Molecule-1 ,Haematopoiesis ,Arterioles ,030104 developmental biology ,Hypoxia-Inducible Factor 1 ,Stem cell ,Signal Transduction - Abstract
Blood vessels define local microenvironments in the skeletal system, play crucial roles in osteogenesis and provide niches for haematopoietic stem cells. The properties of niche-forming vessels and their changes in the ageing organism remain incompletely understood. Here we show that Notch signalling in endothelial cells leads to the expansion of haematopoietic stem cell niches in bone, which involves increases in CD31-positive capillaries and platelet-derived growth factor receptor-β (PDGFRβ)-positive perivascular cells, arteriole formation and elevated levels of cellular stem cell factor. Although endothelial hypoxia-inducible factor signalling promotes some of these changes, it fails to enhance vascular niche function because of a lack of arterialization and expansion of PDGFRβ-positive cells. In ageing mice, niche-forming vessels in the skeletal system are strongly reduced but can be restored by activation of endothelial Notch signalling. These findings indicate that vascular niches for haematopoietic stem cells are part of complex, age-dependent microenvironments involving multiple cell populations and vessel subtypes.
- Published
- 2015
31. Mitochondria Transfer from Hematopoietic Stem and Progenitor Cells to Pdgfrα+/Sca-1-/CD48dim BM Stromal Cells Via CX43 Gap Junctions and AMPK Signaling Inversely Regulate ROS Generation in Both Cell Populations
- Author
-
Karin Golan, Jose A. Cancelas, Toshio Suda, Tao Cheng, Eman Khatib-Massalha, Orit Kollet, Mark J Althoff, Yuji Takihara, Ashley M Wellendorf, Marie-Dominique Fillipi, Tsvee Lapidot, Shiri Gur-Cohen, Tomer Itkin, Hui Cheng, and Anju Kumari
- Subjects
0301 basic medicine ,Stromal cell ,biology ,Chemistry ,Immunology ,Cell ,AMPK ,Cell Biology ,Hematology ,Mitochondrion ,Biochemistry ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,biology.protein ,Stromal cell-derived factor 1 ,Stem cell ,Progenitor cell - Abstract
Modulation of reactive oxygen species (ROS) levels in hematopoietic stem cells (HSC) is crucial to control HSC quiescence and blood formation. High ROS levels are required for leukocyte formation while low ROS levels are essential to maintain HSC quiescence. However, regulation of ROS content in HSC is poorly understood. Adhesion interactions between HSC and their bone marrow (BM) stromal cells (BMSC) via CXCL12/CXCR4 maintain HSC in a quiescence non-motile state, protecting them from 5-FU chemotherapy insult (Sugiyama, Immunity, 2006). Surface CXCL12 expression by BMSC is dependent on connexin-43 (Cx43) gap junctions mediated cell contact (Schajnovitz, Nat. Immunol., 2011) and BM hematopoietic stem and progenitor cells (HSPC) survive and eliminate excess ROS levels post 5-FU chemotherapy treatment, by transferring ROS to BMSC in a Cx43 dependent manner (Taniguchi, PNAS 2012). Here, we report that ROS content of BM HSPC inversely correlates with ROS levels in adjacent BMSC. Administration of the pro inflammatory cytokine G-CSF results in decreased HSPC Cx43 expression, elevated ROS levels and increased glucose uptake. Conversely, in the BM stromal microenvironment, G-CSF administration generated lower ROS level and reduced glucose uptake. Up-regulation of BM Sphingosine 1-Phosphate (S1P), a downstream target of G-CSF required for ROS production in HSPC, reduced stromal ROS content and proliferation. Accordingly, mice with reduced BM S1P levels (S1Plow) have lower BM content of HSPC, accompanied by reduced ROS, glucose uptake and lactate production in these cells. More importantly, BM from S1Plow mice has a 3 fold increased frequency of primitive ROSlow/ EPCR+ long-term repopulating cells, as evident by immunophenotypic analysis and long-term competitive repopulation assays. Concomitantly, S1Plow mice have increased content of BMSC with higher ROS levels and glucose uptake, leading to higher BM content of colony-forming unit fibroblasts. Our results reveal a dynamic and inverse metabolic relationship between BM HSC and the stroma microenvironment. We hypothesized that the opposite metabolic state of HSPC and BMSC is due to mitochondrial transfer between the two populations. Therefore, we created chimeric mice by transplanting mitochondria labeled GFP (mito-GFP) HSPC to wild type (WT) mice and detected 88% of the host BMSC to contain donor-derived mitochondria, indicating the existence of mitochondria transfer from hematopoietic cells to BMSC in vivo. This transfer is bidirectional, albeit at a lesser degree, as determined in reverse chimeric mice where up to 26% of the donor-derived HSPCs acquired recipient mitochondria. Mitochondrial transfer can be recapitulated also in vitro in an overnight co-culture system of mito-GFP HSPC and primary BMSC, resulting in mitochondrial transfer and increased ROS content in a subpopulation of osteogenic BM PDGFRα+/ Sca-1-/CD48dim stromal cells. Mitochondrial transfer is cell contact dependent and mediated by Cx43 gap junctions. In vitro co-culture of mito-GFPHSPC from Cx43 deficient (KO) mice with WT or Cx43 KO BMSC reduced 50% mitochondrial transfer to PDGFRα+/Sca-1-/CD48dim stromal cells. Contrarily, the mitochondrial transfer from WT HSPC to Cx43 KO stromal cells was not affected, revealing that Cx43 expression on HSPC, but not on BM stromal cells, is specifically required for mitochondrial transfer. Interestingly, in vitro inhibition of AMP-activated protein kinase (AMPK), a crucial metabolic regulator, dramatically increased mitochondrial transfer from HSPC to BMSC. Administration of the AMPK inhibitor BML in vivo increased ROS content of PDGFRα+/Sca-1- BMSC while decreasing it in HSPC, further suggesting that AMPK inhibition regulates mitochondrial transfer and ROS production. Our results imply that mitochondria are scavenged by the BM osteogenic microenvironment to prevent excessive ROS levels in the HSC pool and in parallel to activate bone formation. Altogether, we have discovered a dynamic, inverse metabolic state between BM HSPC and their supporting stromal microenvironment during quiescence, proliferationand differentiation of these two populations. Thus, blood cell production and bone generation take place at the expense of the other. This metabolic seesaw is mediated by mitochondrial transfer from HSPC to osteogenic BM stroma in a HSPC Cx43 gap-junction dependent manner and regulated through AMPK signaling. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
32. Inverse PAR1 Activity of Hematopoietic Stem Cells and BM Stromal Cells Mediates G-CSF-Induced Mobilization By Regulation of Nitric Oxide Generation
- Author
-
Benjamin Brenner, Seymen Avci, Shiri Gur-Cohen, Wolfram Ruf, T. Zuckerman, Orit Kollet, Myriam Weyl Ben Arush, Tsvee Lapidot, Yona Nadir, Charles T. Esmon, Neta Nevo, Irina Zaidman, Tomer Itkin, Francesca Avemaria, and Sagarika Chakrabarty
- Subjects
0301 basic medicine ,Stromal cell ,biology ,Chemistry ,Immunology ,Mesenchymal stem cell ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biochemistry ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,biology.protein ,Stromal cell-derived factor 1 ,Progenitor cell ,Stem cell ,Hematopoietic Stem Cell Mobilization - Abstract
Hematopoietic stem and progenitor cell (HSPC) egress from the bone marrow (BM) to the circulation is tightly regulated and is accelerated during stress conditions, a process utilized for BM harvest. Recently, we demonstrated that mouse long term repopulating hematopoietic stem cell (LT-HSC) BM retention and their rapid release to the blood circulation are governed by a switch in nitric oxide (NO) generation via distinct coagulation-related protease activated receptor 1 (PAR1) cascades (Gur-Cohen S. et al., NM, 2016). Herein we report that surface PAR1 expression can be exploited and serve as a positive predictive marker for the efficiency of human CD34+ HSPC mobilization among healthy donors in clinical G-CSF-induced mobilization for matched allogeneic transplantations. We found that PAR1 expression on circulating leukocytes before G-CSF administration was positively correlated with higher yields of mobilized leukocytes after stimulation (P0.5 x 109/L) from an average of 14.6 days to an average of 11 days post-transplant. In addition, a trend of accelerated platelet production was documented to be related with higher PAR1 expression by circulating leukocytes prior to G-CSF stimulation. Consequently, poor mobilizer donors were characterized by extremely low surface PAR1 expression on circulating CD34+ cells prior to G-CSF stimulations. Herein we present a case report of a thrombophilic donor carrying the MTHFR mutation, expressing exceedingly low PAR1 levels at baseline, with the outcome of inadequate numbers of mobilized CD34+ HSPC in the blood following G-CSF treatments. To further gain insight into the role played by PAR1 signaling in the regulation of G-CSF-induced HSPC mobilization, we used mice as a functional preclinical small animal model. We found that antagonizing PAR1 signaling attenuated both steady state release and G-CSF-induced HSPC mobilization. Furthermore, co-administration of G-CSF with PAR1 antagonist attenuated secretion of BM stromal CXCL12 and abrogated upregulation of surface CXCR4 and PAR1 expression by BM HSPCs, all leading to significantly reduced HSPC migration, differentiation and mobilization. In support, PAR1-/- mice failed to efficiently mobilize HSPCs in response to G-CSF compared to wild type counterparts. Enforced HSPC recruitment by G-CSF treatments dramatically accelerated PAR1-dependent NO production by eNOS, known to promote TACE-mediated EPCR shedding and rapid LT-HSC mobilization. Concomitantly, circulating steady state and G-CSF-mobilized stem cells lack surface EPCR expression. Intriguingly, while EPCR expression by primitive BM stem cells was transiently reduced after G-CSF treatments, antagonizing PAR1 signaling along with G-CSF stimuli blocked NO generation and synchronically expanded BM EPCR+ LT-HSC and their supportive stromal progenitor cells (MSPCs), as confirmed by increased repopulation in transplanted mice. Finally, we report an inverse PAR1 expression and regulation by BM HSPC and stromal MSPCs in mediating G-CSF-induced mobilization. G-CSF induced elevation of PAR1 expression on BM HSPCs, providing the driving force for their enhanced NO mediated migration, proliferation, differentiation and recruitment to the circulation. Contrary, the levels of PAR1 expression were reduced on MSPCs in response to G-CSF treatment, and following NO generation by eNOS activity and CXCL12 secretion, resulted in reduced mesenchymal differentiation leading to accumulated numbers of immature mesenchymal (CFU-F) and osteoblast (CFU-OB) progenitor cells. Taken together, our study identifies and highlights inverse PAR1 signaling and NO generation as essential regulator of G-CSF induced HSPC mobilization and MSPC development opening new avenues to advance therapeutics for enhancing clinical G-CSF induced stem cell mobilization and transplantation protocols. Disclosures Ruf: Iconic Therapeutics: Consultancy.
- Published
- 2016
33. Daily Light and Darkness Signals Regulate Bone Marrow Stem Cell Development and Leukocyte Production Via Tnfα and an Interplay Between Norepinephrine and Melatonin
- Author
-
Karin Golan, Regina P. Markus, Anju Kumari, Sylwia Rzeszotek, Mariusz Z. Ratajczak, Tsvee Lapidot, Tomer Itkin, Shiri Gur-Cohen, Orit Kollet, Zulma S. Ferreira, and Eman Khatib-Massalha
- Subjects
education.field_of_study ,Chemistry ,Cellular differentiation ,Monocyte ,Immunology ,Population ,Bone Marrow Stem Cell ,Cell Biology ,Hematology ,Biochemistry ,Cell biology ,Melatonin ,Blood cell ,medicine.anatomical_structure ,medicine ,Stem cell ,Progenitor cell ,education ,medicine.drug - Abstract
How bone marrow (BM) stem cells replenish the blood with mature cells while maintaining the reservoir of undifferentiated stem cells, is poorly understood. We report that murine leukocyte production and BM stem cell maintenance are regulated by light and darkness cues. We identified two daily peaks of BM stem and progenitor cell (HSPC) proliferation: the morning peak following light initiation (11 AM, ZT5) and the night peak following darkness (11 PM, ZT17). Both peaks are preceded by a transient elevation of tumor necrosis factor-alpha (TNFα) in the BM at 7 AM and at 7 PM, leading to increased reactive oxygen species (ROS) in HSPC and inducing their cycling. Reduced HSPC levels were observed either following ROS inhibition or in TNFα deficient mice. TNFα elevation augmented the levels of the TNFα converting enzyme (TACE) levels on HSPCs, promoting BM TNFα shedding. Interestingly, transient TNFα elevation was induced by switching light to darkness and vice versa, suggesting a role for TNFα as an internal mechanism of alert, preparing HSPC to cycle upon demand. While the morning HSPC peak was accompanied by increased egress and differentiation, the night peak was associated with retention and low differentiation. Norepinephrine (NE) generation has been shown to be driven by light-induced cues from the brain and to induce stem cell egress from the BM during the morning peak (Mendez-Ferrer et al, Nature 2008), while melatonin is an antioxidant that is mainly produced following the onset of darkness. We found that although NE and melatonin are continuously present in the BM, NE levels are predominantly augmented following initiation of light while melatonin is mostly elevated during the night. Administration of melatonin or inhibition of the sympathetic nervous system by β3-adrenergic receptor antagonist during the morning induced HSPC retention, decreasing their morning differentiation and egress. In accordance, injection of NE during the evening induced HSPC egress and differentiation at night. Taken together, these results reveal that TNFα via ROS generation regulates both light and darkness peaks of stem cell proliferation in the BM. However, the nervous system via NE secretion further drives their maturation and egress only during the morning peak. Looking for mechanisms of HSPC protection which are essential to avoid BM exhaustion, we found that melatonin prevented their differentiation and egress thus maintaining them in a primitive state during the darkness peak. Concomitant with the night peak, we also observed increased BM levels of rare activated αSMA/Mac-1 macrophage/monocyte cells. This population maintains HSPC in a primitive state via COX2/PGE2 signaling that reduces ROS levels and increases BM stromal CXCL12 surface expression (Ludin et al, Nat. Imm. 2012). The high melatonin levels at night induced PGE2 signaling in the BM stem cell niche, regulating COX2high αSMA/Mac-1 macrophages, which restored low ROS levels, preventing stem cell differentiation and egress. Murine BM leukocytes differentiate predominantly during the light time and are therefore more responsive to inflammatory challenges during this time frame. Mimicking bacterial infections, endotoxin-induced mortality was shown to correlate with administration time, with very high mortality in mice treated at noon and very low mortality following midnight challenge (Halberg et al, Exp boil Med, 1960). We found that LPS administration in the afternoon resulted in a dramatic increase in BM neutrophils and monocytes production and recruitment which is lethal, in contrast to LPS injection at midnight with no immune activation. Reducing differentiation in the BM during the morning peak by administrating β3-adrenergic receptor antagonist, melatonin or ROS inhibition, all decreased the levels of myeloid cell production and recruitment following LPS challenge in the afternoon. Our results revealed that the morning peak involves HSPC proliferation, differentiation and egress, allowing HSPC to replenish the blood and the immune system with mature leukocytes on a daily basis. In contrast, the night peak induces HSPC proliferation with reduced differentiation and egress, allowing the renewal of the BM stem cell pool. In summary, we have identified two daily peaks in BM HSPC levels which are regulated via light and darkness cues that impact daily blood cell production, host immunity and renewal of the BM stem cell reservoir. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
34. EPCR Guides Hematopoietic Stem Cells Homing to the Bone Marrow Independently of Niche Clearance
- Author
-
Wolfram Ruf, Mayela M. Carolina, Tsvee Lapidot, Eman Khatib-Massalha, Qing-Cissy Yu, Hua Lin Wu, Charles T. Esmon, Seymen Avci, Orit Kollet, Anjali P. Kusumbe, Ralf H. Adams, Elizabeth J. Shpall, Francesca Avemaria, Ayelet Erez, Shiri Gur-Cohen, Wei-Ling Lin, Anju Kumari, Tomer Itkin, Conway M. Edward, Saravana K. Ramasamy, Yi Arial Zeng, Irit Sagi, and Karin Golan
- Subjects
Endothelial protein C receptor ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Cancer stem cell ,medicine ,Bone marrow ,Progenitor cell ,Stem cell ,Homing (hematopoietic) - Abstract
Bone marrow (BM) homing and lodgment of long-term repopulating hematopoietic stem cells (LT-HSCs) are active and essential first steps during embryonic development and in clinical stem cell transplantation. Rare, BM LT-HSCs endowed with the highest self-renewal and durable repopulation potential, functionally express the anticoagulant endothelial protein C receptor (EPCR) and PAR1. In addition to coagulation and inflammation, EPCR-PAR1 signaling independently controls a BM LT-HSC retention-release switch via regulation of nitric oxide (NO) production within LT-HSCs. EPCR+ LT-HSCs are maintained in thrombomodulin+ (TM) periarterial BM microenvironments via production of activated protein C (aPC), the major ligand for EPCR. Restriction of NO production by aPC-EPCR-PAR1 signaling, activates VLA4-mediated adhesion, anchoring EPCR+ LT-HSCs to the BM and protecting them from chemotherapy insult, sparing hematological failure and premature death (Gur-Cohen S. et al, Nat Med 2015). We report that transplanted EPCR+ LT-HSCs preferentially homed to and were retained in the BM, while immature progenitors were equally distributed between the BM and spleen. Specificity of BM homing was further confirmed by EPCR neutralizing treatment that block aPC binding and attenuate EPCR+ LT-HSC BM homing. Furthermore, short term aPC in vitro pretreatment dramatically augmented EPCR+ LT-HSC BM homing, lodgment and long-term repopulation. PAR1 deficient stem cells were irresponsive to treatment with aPC and displayed reduced BM homing efficiency, all pointing to the aPC-EPCR-PAR1 axis as a crucial mediator of BM LT-HSC homing. Additionally, aPC pretreated EPCR+ LT-HSCs had a striking advantage to competitively home to the BM. Consistently, BM HSCs obtained from Procrlow mice, expressing markedly reduced surface EPCR, failed to compete with wild type stem cells in competitive repopulation assays. Importantly, the competitive homing results strongly imply that the BM available niches for newly arrived EPCR+ LT-HSCs are limited. Indeed, aPC pretreated EPCR+ LT-HSCs BM homing reached a plateau, as increasing the transplanted cell dose above 5x106 BM mononuclear cells, did not yield higher donor EPCR+ LT-HSC homing. These results reveal that there is a limited BM space for newly arrived transplanted EPCR+ stem cells to non-irradiated hosts. Importantly, we found that EPCR+ LT-HSCs can engraft the BM of non-conditioned mice with high efficiency, while remaining in a dormant, non-cycling state. Furthermore, the dormant homed EPCR+ LT-HSCs were later awakened and activated solely by treating the engrafted hosts with a low dose 5-FU chemotherapy, or with NO donor SNAP, revealing that preconditioning and clearance of occupied BM HSC niches are not required. To further address the preferential homing of EPCR+ LT-HSCs to the BM, we found that TM is exclusively expressed by unique BM arterioles, and not in the spleen. BM homed EPCR+ LT-HSCs were found adjacent to TM+ arterioles, imposing their retention. Homed BM EPCR+ LT-HSCs highly express full-length TM with intact lectin-like domain, and the BM TM+ endothelium was found to be enriched with a Glycocalyx layer, in particular with Heparan Sulfate Proteoglycan-2 (HSPG-2). HSGP-2 might specifically interact with the lectin-like domain of TM-expressingLT-HSCs, providing BM specific recognition and accelerated homing. Intriguingly, stabilizing TM function by in vitro pretreatment with platelet factor-4 (PF4) bypassed BM-derived cues and increased EPCR+/TM+ LT-HSC homing also to the spleen, suggesting a supportive role for PF4, highly secreted by BM megakaryocytes, in guiding EPCR+/TM+ LT-HSCs to the BM. Herein we define EPCR as a guidance molecule, navigating LT-HSC specifically to BM TM+ aPC-secreting blood vessels, allowing stem cell retention and protection from DNA damaging agents. The BM harbors a limited number of available stem cell niches for newly arrived transplanted EPCR+/TM+ LT-HSCs, and in vitro aPC pretreatment dramatically augments EPCR+/TM+ LT-HSC BM homing. Our findings provide new mechanistic insights and identify key players concerning LT-HSC homing specifically to the BM, leading to better repopulation following transplantation. This up-to-date approach and new knowledge may potentially lead to improved BM transplantation protocols and to prevent chemotherapy resistance of EPCR-expressing cancer stem cell mediated relapse. Disclosures Ruf: Iconic Therapeutics: Consultancy.
- Published
- 2016
35. Fibroblast growth factor signaling promotes physiological bone remodeling and stem cell self-renewal
- Author
-
Tsvee Lapidot, Tomer Itkin, Aya Ludin, Shiri Gur-Cohen, and Kerstin B. Kaufmann
- Subjects
Tumor microenvironment ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Hematology ,Biology ,Fibroblast growth factor ,Hematopoietic Stem Cells ,Stem Cell Self-Renewal ,Cell biology ,Fibroblast Growth Factors ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Tumor Microenvironment ,Humans ,Bone marrow ,Bone Remodeling ,Progenitor cell ,Stem cell ,Signal Transduction - Abstract
Purpose of review Fibroblast growth factor (FGF) signaling activates many bone marrow cell types, including various stem cells, osteoblasts, and osteoclasts. However, the role of FGF signaling in regulation of normal and leukemic stem cells is poorly understood. This review highlights the physiological roles of FGF signaling in regulating bone marrow mesenchymal and hematopoietic stem and progenitor cells (MSPCs and HSPCs) and their dynamic microenvironment. In addition, this review summarizes the recent studies which provide an overview of FGF-activated mechanisms regulating physiological stem cell maintenance, self-renewal, and motility. Recent findings Current results indicate that partial deficiencies in FGF signaling lead to mild defects in hematopoiesis and bone remodeling. However, FGF signaling was shown to be crucial for stem cell self-renewal and for proper hematopoietic poststress recovery. FGF signaling activation was shown to be important also for rapid AMD3100 or post 5-fluorouracil-induced HSPC mobilization. In vivo, FGF-2 administration successfully expanded both MSPCs and HSPCs. FGF-induced expansion was characterized by enhanced HSPC cycling without further exhaustion of the stem cell pool. In addition, FGF signaling expands and remodels the supportive MSPC niche cells. Finally, FGF signaling is constitutively activated in many leukemias, suggesting that malignant HSPCs exploit this pathway for their constant expansion and for remodeling a malignant-supportive microenvironment. Summary The summarized studies, concerning regulation of stem cells and their microenvironment, suggest that FGF signaling manipulation can serve to improve current clinical stem cell mobilization and transplantation protocols. In addition, it may help to develop therapies specifically targeting leukemic stem cells and their supportive microenvironment.
- Published
- 2013
36. GSK3β regulates physiological migration of stem/progenitor cells via cytoskeletal rearrangement
- Author
-
Karin Golan, Giulia Caglio, Gabriele D'Uva, Ziv Porat, Aya Ludin, Tomer Itkin, Yossi Ovadya, Kfir Lapid, Alexander Kalinkovich, Tsvee Lapidot, Massimo Zollo, Lapid K., Itkin T., D'Uva G., Ovadya Y., Ludin A., Caglio G., Kalinkovich A., Golan K., Porat Z., Zollo M., Lapidot T., Lapid, K, Itkin, T, D'Uva, G, Ovadya, Y, Ludin, A, Caglio, G, Kalinkovich, A, Golan, K, Porat, Z, Zollo, Massimo, and Lapidot, T.
- Subjects
Receptors, CXCR4 ,Motility ,Protein Kinase Inhibitor ,Stem cell factor ,Microtubule ,Mice, SCID ,Biology ,Microtubules ,Receptor, IGF Type 1 ,03 medical and health sciences ,Glycogen Synthase Kinase 3 ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,Animals ,Humans ,Progenitor cell ,Insulin-Like Growth Factor I ,GSK3B ,Protein Kinase Inhibitors ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Stem Cell Factor ,Glycogen Synthase Kinase 3 beta ,Animal ,Chemotaxis ,GSK3 beta ,Hematopoietic Stem Cell ,General Medicine ,Actin cytoskeleton ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Cell biology ,Enzyme Activation ,Mice, Inbred C57BL ,Haematopoiesis ,Actin Cytoskeleton ,030220 oncology & carcinogenesis ,Stem cell ,Corrigendum ,Research Article ,Human ,Signal Transduction - Abstract
Regulation of hematopoietic stem and progenitor cell (HSPC) steady-state egress from the bone marrow (BM) to the circulation is poorly understood. While glycogen synthase kinase-3β (GSK3β) is known to participate in HSPC proliferation, we revealed an unexpected role in the preferential regulation of CXCL12-induced migra- tion and steady-state egress of murine HSPCs, including long-term repopulating HSCs, over mature leuko- cytes. HSPC egress, regulated by circadian rhythms of CXCL12 and CXCR4 levels, correlated with dynamic expression of GSK3β in the BM. Nevertheless, GSK3β signaling was CXCL12/CXCR4 independent, suggesting that synchronization of both pathways is required for HSPC motility. Chemotaxis of HSPCs expressing higher levels of GSK3β compared with mature cells was selectively enhanced by stem cell factor–induced activation of GSK3β. Moreover, HSPC motility was regulated by norepinephrine and insulin-like growth factor-1 (IGF-1), which increased or reduced, respectively, GSK3β expression in BM HSPCs and their subsequent egress. Mech- anistically, GSK3β signaling promoted preferential HSPC migration by regulating actin rearrangement and microtubuli turnover, including CXCL12-induced actin polarization and polymerization. Our study identifies a previously unknown role for GSK3β in physiological HSPC motility, dictating an active, rather than a pas- sive, nature for homeostatic egress from the BM reservoir to the blood circulation.
- Published
- 2013
37. Correction: Corrigendum: Age-dependent modulation of vascular niches for haematopoietic stem cells
- Author
-
Ralf H. Adams, Anjali P. Kusumbe, Tomer Itkin, Christer Betsholtz, Urs H. Langen, Maarja Mäe, Tsvee Lapidot, and Saravana K. Ramasamy
- Subjects
Haematopoiesis ,Molecular interactions ,Multidisciplinary ,Age dependent ,Computational biology ,Stem cell ,Biology - Abstract
Nature 532, 380–384 (2016); doi:10.1038/nature17638 In this Letter, the graph for the Fbxw7 mutants in Fig. 2e was a duplicate of the NICD graph. This inadvertently occurred during the reformatting of the accepted manuscript, and the panel has now been corrected in Fig. 1 of this Corrigendum, but has not been corrected in the online versions of the original paper.
- Published
- 2016
38. Erratum: Corrigendum: Distinct bone marrow blood vessels differentially regulate haematopoiesis
- Author
-
Anjali P. Kusumbe, Anju Kumari, Aya Ludin, Karin Golan, Yookyung Jung, Shahin Rafii, Eman Khatib, Shiri Gur-Cohen, Alexander Kalinkovich, Michael G. Poulos, David T. Scadden, Joel A. Spencer, Guy Ledergor, Tsvee Lapidot, Orit Kollet, Ralf H. Adams, Tomer Itkin, Amir Schajnovitz, Idan Milo, Charles P. Lin, Jason M. Butler, Guy Shakhar, and Saravana K. Ramasamy
- Subjects
0301 basic medicine ,03 medical and health sciences ,Haematopoiesis ,Multidisciplinary ,medicine.anatomical_structure ,030102 biochemistry & molecular biology ,Section (typography) ,medicine ,Bone marrow ,Anatomy ,Biology ,Bioinformatics ,Stem cell niche - Abstract
Nature 532, 323–328 (2016); doi:10.1038/nature17624 The authors Karin Golan, Eman Khatib and Anju Kumari were erroneously omitted from the author list of this Article. They are all associated with the affiliation: Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel, and the Author Contributions section should have included the following statements: K.
- Published
- 2016
39. Erratum: Corrigendum: PAR1 signaling regulates the retention and recruitment of EPCR-expressing bone marrow hematopoietic stem cells
- Author
-
Orit Kollet, Shiri Gur-Cohen, Alexander Kalinkovich, Wolfram Ruf, Sagarika Chakrabarty, Tsvee Lapidot, Ziv Porat, Karin Golan, Charles T. Esmon, Eitan Wong, Guy Ledergor, Ayelet Erez, Elisabeth Niemeyer, Tomer Itkin, Irit Sagi, Claudine Graf, and Aya Ludin
- Subjects
0301 basic medicine ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,business.industry ,Cancer research ,Medicine ,General Medicine ,Bone marrow ,Stem cell ,business ,General Biochemistry, Genetics and Molecular Biology - Abstract
Nat. Med. 21, 1307–1317 (2015); published online 12 October 2015; corrected after print 18 November 2015 In the version of this article initially published, the first author's name was incorrect. The error has been corrected in the HTML and PDF versions of the article.
- Published
- 2016
40. S1P promotes murine progenitor cell egress and mobilization via S1P1-mediated ROS signaling and SDF-1 release
- Author
-
Orit Kollet, Shiri Cohen-Gur, Shoham Shivtiel, Andrew J. Morris, Alexander Kalinkovich, Amir Schajnovitz, Tsvee Lapidot, Kfir Lapid, Mariusz Z. Ratajczak, Aya Ludin, Chihwa Kim, Yossi Ovadya, Tomer Itkin, Yaron Vagima, and Karin Golan
- Subjects
Male ,Benzylamines ,Stromal cell ,Hematopoiesis and Stem Cells ,Immunology ,Fluorescent Antibody Technique ,Biology ,Cyclams ,Biochemistry ,Colony-Forming Units Assay ,Mice ,Bone Marrow ,Cell Movement ,Heterocyclic Compounds ,Sphingosine ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Progenitor cell ,Hematopoietic Stem Cell Mobilization ,Cells, Cultured ,Mice, Knockout ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Cell biology ,Endothelial stem cell ,Mice, Inbred C57BL ,Haematopoiesis ,Phosphotransferases (Alcohol Group Acceptor) ,Receptors, Lysosphingolipid ,medicine.anatomical_structure ,Female ,Bone marrow ,Stem cell ,Lysophospholipids ,Stromal Cells ,Reactive Oxygen Species ,Signal Transduction - Abstract
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 S1P1, 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 S1P1 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/S1P1 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.
- Published
- 2012
41. Monocytes-macrophages that express α-smooth muscle actin preserve primitive hematopoietic cells in the bone marrow
- Author
-
Orit Kollet, Alexander Mildner, Karin Golan, Tomer Itkin, Ron N. Apte, Elias Shezen, David A. Brenner, Elena Voronov, Ziv Porat, Steffen Jung, Tsvee Lapidot, Aya Ludin, Gabriele D'Uva, Amir Schajnovitz, Shiri Gur-Cohen, Alexander Kalinkovich, Ludin A., Itkin T., Gur-Cohen S., Mildner A., Shezen E., Golan K., Kollet O., Kalinkovich A., Porat Z., D'Uva G., Schajnovitz A., Voronov E., Brenner D.A., Apte R.N., Jung S., and Lapidot T.
- Subjects
Cell Survival ,Macrophage ,Immunology ,Clinical uses of mesenchymal stem cells ,Cell Communication ,Biology ,Monocyte ,CXCR4 ,Monocytes ,Dinoprostone ,Blood cell ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Bone Marrow ,Cell Movement ,medicine ,Immunology and Allergy ,Animals ,Actin ,030304 developmental biology ,0303 health sciences ,Animal ,Macrophages ,Gamma Ray ,Mesenchymal Stem Cells ,Hematopoietic Stem Cell ,Hematopoietic Stem Cells ,Actins ,Chemokine CXCL12 ,3. Good health ,Cell biology ,Endothelial stem cell ,Haematopoiesis ,medicine.anatomical_structure ,Mesenchymal Stem Cell ,Gene Expression Regulation ,Gamma Rays ,Cyclooxygenase 2 ,030220 oncology & carcinogenesis ,Bone marrow ,Stem cell ,Reactive Oxygen Species ,Reactive Oxygen Specie ,Proto-Oncogene Proteins c-akt ,Adult stem cell ,Signal Transduction - Abstract
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 E 2 (PGE 2) 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. © 2012 Nature America, Inc. All rights reserved.
- Published
- 2012
42. Distinct Bone Marrow Blood Vessels Differentially Regulate Normal and Malignant Hematopoietic Stem and Progenitor Cells
- Author
-
David T. Scadden, Shahin Rafii, Aya Ludin, Joel A. Spencer, Shiri Gur Cohen, Saravana K. Ramasamy, Guy Ledergor, Alexander Kalinkovich, Tomer Itkin, Amir Schajnovitz, Guy Shakhar, Simón Méndez-Ferrer, Jason M. Butler, Charles P. Lin, Michael G. Poulos, Tsvee Lapidot, Anjali P. Kusumbe, Orit Kollet, Ralf H. Adams, Maria Garcia Fernandez, Yookyung Jung, and Idan Milo
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,Endothelium ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Leukocyte Trafficking ,medicine ,biology.protein ,Stromal cell-derived factor 1 ,Bone marrow ,Progenitor cell ,Stem cell - Abstract
Bone marrow (BM) endothelial cells (BMECs) form a network of blood vessels (BVs) that regulate both leukocyte trafficking and hematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance between these dual regulatory roles and if these events occur at the same vascular site. We define the BM architecture of functionally distinct BVs, their spatial localization and association with specific stromal precursors, which cooperatively regulate HSPC fate. BM stem and progenitor cell maintenance in a metabolically non-active state and leukocyte trafficking occur at separate sites and are differentially regulated by specific BVs with distinct permeability properties. BM arteries were found to be mostly encircled by aSMA+ pericytes whereas the ensuing small-diameter endosteal and trabecular arterioles were predominantly surrounded by stem cell-niche supporting stromal precursor cells. Live imaging and flow analysis revealed that endosteal arteriole BVs exhibited high flow rate, low permeability to external plasma from the peripheral blood, and high levels of adhesion- and tight-junction molecules. Primitive HSPCs located in peri-arteriole regions were found in a non-activated, low reactive oxygen species (ROS) state. Exposure of BM HSPCs to peripheral blood plasma, enhanced their metabolic activity, exhibited by enhanced intracellular ROS levels, and glucose uptake. The same was also evident for circulating HSPCs in the blood. Plasma-exposed HSPCs displayed enhanced motility alongside with reduced long-term repopulation potential. Live imaging showed that all immature and mature leukocyte bi-directional trafficking occurred exclusively at the more permeable sinusoids, located downstream to the endosteal arterioles. Of note, BM sinusoids contained a higher prevalence of ROShigh cells in their microenvironment, including HSPCs. Rapid AMD3100-induced HSPC mobilization preferentially affected sinusoidal but not arterial BVs permeability and CXCL12 chemokine release. Endothelial specific in vivo interference with CXCL12-CXCR4 interactions, via conditional CXCR4 genetic deletion, hampered BM barrier integrity resulting in enhanced HSPC egress. In line with these results we found that during conditions favoring BM stem and progenitor cells expansion, endothelial integrity was enhanced along with reduced HSPC bi-directional trafficking. Conversely, conditional endothelial specific induced genetic or pharmacologic disruption of barrier integrity augmented ROS levels in HSPCs, enhancing their bi-directional trafficking and differentiation while reducing their BM pool size and maintenance in a metabolically non-active state. Of note, humanized mice engrafted with pre-B ALL cells exhibited reduced BM barrier permeability most probably due to BM endothelium modification via FGF-2 secretion by the leukemic clone. Interestingly, human pre-B ALL cells displayed hypersensitivity to plasmatic exposure. We hypothesize that malignant cells modify BM endothelium to provide themselves with a supportive and protective microenvironment composed of undifferentiated BM stromal progenitors and tightly sealed endothelial barrier. In conclusion, our study identifies anatomically distinct BM BVs with different barrier functions serving as systemic leukocyte trafficking or HSPC BM maintenance sites with clinical therapeutic relevance. Disclosures Rafii: Angiocrine Bioscience: Consultancy, Equity Ownership.
- Published
- 2015
43. EPCR/PAR1 Signaling Navigates Long-Term Repopulating Hematopoietic Stem Cell Bone Marrow Homing to Thrombomodulin-Enriched Blood Vessels
- Author
-
Tsvee Lapidot, Orit Kollet, Alexander Kalinkovich, Karin Golan, Irit Sagi, Claudine Graf, Wolfram Ruf, Ayelet Erez, Charles T. Esmon, Sagarika Chakrabarty, Shiri Gur Cohen, and Tomer Itkin
- Subjects
Immunology ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Transplantation ,Endothelial stem cell ,Haematopoiesis ,medicine.anatomical_structure ,Cdc42 GTP-Binding Protein ,medicine ,Stem cell ,Progenitor cell ,Homing (hematopoietic) - Abstract
Bone marrow (BM) homing and lodgment of long-term repopulating hematopoietic stem cells (LT-HSCs) is an active and essential first step in clinical stem cell transplantation. EPCR is expressed by murine BM LT-HSCs endowed with the highest repopulation potential and its ligand, activated protein C (aPC), has anticoagulant and anti-sepsis effects in EPCR+/PAR1+ endothelial cells. We recently found that signaling cascades, traditionally viewed as coagulation and inflammation related, also independently control EPCR+ LT-HSC BM retention and recruitment to the blood via distinct PAR1 mediated pathways. EPCR/PAR1 signaling retains LT-HSCs in the BM by restricting nitric oxide (NO) production and Cdc42 activity, promoting VLA4 affinity and adhesion. Conversely, thrombin/PAR1 signaling overcome EPCR+ LT-HSC BM retention by initiating NO production, leading to TACE-mediated EPCR shedding, CXCR4 and PAR1 upregulation and parallel CXCL12 secretion by PAR1+ BM stromal cells, enhancing stem cell migration and mobilization. Since EPCR shedding is essential for BM LT-HSC recruitment, we tested EPCR role in LT-HSC BM homing. EPCR+ LT-HSC exhibited reduced in vitro migration towards CXCL12 and enhanced CXCL12-dependent adhesion to fibronectin. Unexpectedly, transplanted EPCR+ LT-HSCs preferentially homed to the host BM, while immature progenitors were equally distributed between the BM and spleen. Specificity of BM homing was further confirmed by EPCR neutralizing antibody treatment, which blocks binding to aPC, leading to attenuated EPCR+ LT-HSC homing to the BM but not to the spleen. Importantly, short term aPC pretreatment inhibited NO production and dramatically increased EPCR+ LT-HSC BM homing. Since EPCR navigates LT-HSC to the BM, we studied the role of EPCR signaling in LT-HSC BM repopulation. Mimicking EPCR signaling by in vivo NO inhibition induced preferential expansion of blood and bone-forming stem cells and gave rise to higher donor type EPCR+ LT-HSCs in competitive repopulation assays. Similarly, repeated treatment with aPC expanded BM EPCR+ stem cells and increased competitive LT-repopulation. Importantly, loss of EPCR function reduced HSC long-term repopulation ability while maintaining their short-term repopulation activity. BM HSCs obtained from Procrlow mice, expressing markedly reduced surface EPCR, failed to compete with normal stem cells in competitive long-term repopulation assays. Consistent with inferior HSC BM repopulation, Procrlow mice exhibited reduced numbers of BM LT-HSC with reduced adhesion capacity. Additionally, these mice displayed increased HSC frequencies in the blood circulation and the spleen, which were pharmacologically corrected by inhibiting NO generation with L-NAME treatment. BM retention is essential for quiescent HSC protection from chemotherapy. Mice treated with NO donor SNAP, or with blocking EPCR antibody as well as Fr2-/-mice lacking PAR1 expression, were more susceptible to hematological failure and mortality induced by 5-FU treatment compared to control mice. Together, these results indicate a functional aPC/EPCR/PAR1 signaling pathway, regulating EPCR+ LT-HSC BM homing, adhesion and long-term repopulation potential. The thrombin-thrombomodulin (TM) complex converts protein C to its activated form aPC, facilitating high affinity binding to its receptor EPCR. To further address the preferential homing of EPCR+ LT-HSCs to the BM, we found that TM is exclusively expressed by a unique BM endothelial cell (BMEC) subpopulation, but not in the spleen. Moreover, EPCR+ LT-HSCs were found adjacent to TM+/aPC+ BMECs, imposing their adhesion and retention. Interestingly, similar to BMECs, BM EPCR+ LT-HSC also express surface TM, implying the possibility of autocrine aPC generation. Herein we define EPCR as a guidance molecule, navigating slow migrating LT-HSC in the blood flow specifically to TM+ BMEC supporting niches, maintaining NOlow stem cell retention, long-term blood production and protection from myelotoxic insult. Conversely, thrombin/PAR1 signaling oppositely increase NO generation and EPCR shedding allowing increased CXCR4-dependent LT-HSC migration and mobilization. Harnessing EPCR signaling may improve clinical stem cell transplantation, increasing LT-HSC specific BM homing and repopulation by aPC pretreatment, as well as potentially to overcome malignant stem cell chemotherapy resistance. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
44. Chronic B cell deficiency from birth prevents age-related alterations in the B lineage
- Author
-
Gitit Shahaf, Karin Golan, Doron Melamed, Zohar Keren, Dana Averbuch, Tsvee Lapidot, Simona Zisman-Rozen, Tomer Itkin, and Ramit Mehr
- Subjects
medicine.medical_specialty ,Aging ,Lineage (genetic) ,Immunology ,Antigens, CD19 ,Receptors, Antigen, B-Cell ,Mice, Transgenic ,Cell Separation ,CD19 ,Mice ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Homeostasis ,Cell Lineage ,Receptor ,B cell ,B-Lymphocytes ,biology ,Lymphopoiesis ,Histocompatibility Antigens Class II ,Cell Differentiation ,Flow Cytometry ,Hematopoietic Stem Cells ,Antigens, Differentiation, B-Lymphocyte ,Mice, Inbred C57BL ,Haematopoiesis ,Endocrinology ,medicine.anatomical_structure ,biology.protein ,Bone marrow ,Stem cell ,B-Cell Activation Factor Receptor - Abstract
Aging is accompanied by a decline in B lymphopoiesis in the bone marrow and accumulation of long-lived B cells in the periphery. The mechanisms underlying these changes are unclear. To explore whether aging in the B lineage is subjected to homeostatic regulation, we used mutant mice bearing chronic B cell deficiency from birth. We show that chronic B cell deficiency from birth, resulting from impaired maturation (CD19−/− and CD74−/−) or reduced survival (baff-r−/−), prevents age-related changes in the B lineage. Thus, frequencies of early and late hematopoietic stem cells, B lymphopoiesis, and the rate of B cell production do not substantially change with age in these mice, as opposed to wild-type mice where kinetic experiments indicate that the output from the bone marrow is impaired. Further, we found that long-lived B cells did not accumulate and peripheral repertoire was not altered with age in these mice. Collectively, our results suggest that aging in the B lineage is not autonomously progressing but subjected to homeostatic regulation.
- Published
- 2011
45. Rapid mobilization of hematopoietic progenitors by AMD3100 and catecholamines is mediated by CXCR4-dependent SDF-1 release from bone marrow stromal cells
- Author
-
Melania Tesio, Tsvee Lapidot, Michela Battista, Andre Larochelle, Eike C. Buss, Hal E. Broxmeyer, Cynthia E. Dunbar, Wei-Ming Kao, Orit Kollet, Amir Schajnovitz, Ayelet Dar, Nobutaka Fujii, Shinya Oishi, Aya Ludin, Alexander Kalinkovich, Raanan Margalit, Paul S. Frenette, Kfir Lapid, Françoise Baleux, Tomer Itkin, and Neta Netzer Cohen
- Subjects
Cancer Research ,Benzylamines ,Receptors, CXCR4 ,Stromal cell ,Bone Marrow Cells ,Biology ,Cyclams ,CXCR4 ,Article ,Mice ,Norepinephrine ,Heterocyclic Compounds ,medicine ,Animals ,Humans ,Hematopoietic Stem Cell Mobilization ,Cells, Cultured ,Mice, Inbred BALB C ,Hematology ,medicine.disease ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Cell biology ,Endothelial stem cell ,Mice, Inbred C57BL ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Oncology ,Immunology ,Bone marrow ,Stem cell ,Stromal Cells - Abstract
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.
- Published
- 2011
46. B-cell depletion reactivates B lymphopoiesis in the BM and rejuvenates the B lineage in aging
- Author
-
Tsvee Lapidot, Karin Golan, Shulamit Naor, Zohar Keren, Tomer Itkin, Yoshiteru Sasaki, Shahar Nussbaum, Doron Melamed, and Marc Schmidt-Supprian
- Subjects
Aging ,Transgene ,Immunology ,Mice, Transgenic ,Biology ,Biochemistry ,Lymphocyte Depletion ,Mice ,Immune system ,Antigen ,Immunity ,Bone Marrow ,medicine ,Animals ,Humans ,Rejuvenation ,Cell Lineage ,Lymphopoiesis ,Progenitor cell ,B-Lymphocytes ,Stem Cells ,Cell Biology ,Hematology ,Immunosenescence ,Antigens, CD20 ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Bone marrow ,B-Cell Activation Factor Receptor - Abstract
Aging is associated with a decline in B-lymphopoiesis in the bone marrow and accumulation of long-lived B cells in the periphery. These changes decrease the body's ability to mount protective antibody responses. We show here that age-related changes in the B lineage are mediated by the accumulating long-lived B cells. Thus, depletion of B cells in old mice was followed by expansion of multipotent primitive progenitors and common lymphoid progenitors, a revival of B-lymphopoiesis in the bone marrow, and generation of a rejuvenated peripheral compartment that enhanced the animal's immune responsiveness to antigenic stimulation. Collectively, our results suggest that immunosenescence in the B-lineage is not irreversible and that depletion of the long-lived B cells in old mice rejuvenates the B-lineage and enhances immune competence.
- Published
- 2011
47. Mobilization of Hematopoietic Stem and Progenitor Cells
- Author
-
Kfir Lapid, Eike C. Buss, Tomer Itkin, and Tsvee Lapidot
- Subjects
Endothelial stem cell ,Haematopoiesis ,Mobilization ,Biology ,Stem cell ,Progenitor cell ,Cell biology ,Adult stem cell - Published
- 2010
48. SDF-1 keeps HSC quiescent at home
- Author
-
Tomer Itkin and Tsvee Lapidot
- Subjects
Chemokine ,Stromal cell ,biology ,Immunology ,biology.protein ,Stromal cell-derived factor 1 ,Cell Biology ,Hematology ,Receptor ,Biochemistry ,CXCR4 ,Cell biology - Abstract
In this issue of Blood , Tzeng et al report on the generation of adult, conditional SDF-1–deficient mice in which HSC quiescence and endosteal niche localization are impaired.[1][1] The chemokine stromal cell–derived factor 1 (SDF-1; CXCL12), and its major receptor CXCR4, regulate many aspects
- Published
- 2011
49. EPCR Limits Nitric Oxide Levels, Mediating Human and Murine Stem Cell Adhesion and Retention In The Bone Marrow, By Conjugating PAR1 and CXCR4 Signaling
- Author
-
Aya Ludin, Ayelet Erez, Wolfram Ruf, Orit Kollet, Charles T. Esmon, Tsvee Lapidot, Alexander Kalinkovich, Shiri Gur Cohen, Eitan Wong, Karin Golan, Tomer Itkin, Sagarika Chakrabarty, and Irit Sagi
- Subjects
Endothelial protein C receptor ,Immunology ,Proteolytic enzymes ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CXCR4 ,Molecular biology ,Cell biology ,Transplantation ,Haematopoiesis ,Progenitor cell ,Stem cell ,Hematopoietic Stem Cell Mobilization - Abstract
Long term repopulating hematopoietic stem cells (LTR-HSC) in the murine bone marrow (BM) highly express endothelial protein C receptor (EPCR), yet the function of EPCR in HSC is incompletely defined. EPCR is expressed primarily on endothelial cells and has anti coagulation and anti inflammatory roles. While physiological stress due to injury or bleeding is a strong inducer of HSC mobilization and leukocyte production, a role for the coagulation protease thrombin, and its major receptor PAR1 in regulation of HSC has not yet been identified. We hypothesized that thrombin plays a role in HSC mobilization in the context of injury and that, conversely, signaling involving EPCR and its ligand activated protein C (aPC) play a regulatory role in HSC maintenance. Herein, we report that murine BM EPCRhigh stem cells display enhanced CXCL12 mediated adhesion and reduced migration capacitie, while motile circulating HSC in the murine blood and spleen lack high EPCR expression. Mechanistically, we found that EPCR is a negative regulator of nitric oxide (NO) levels. EPCRhigh stem cells display low intracellular NO levels, low motility, and increased adhesion to BM stroma. Furthermore, EPCRlow transgenic mouse cells displayed reduced stem cell adhesion to BM stroma and increased motility, manifested by reduced EPCRlow HSC in the BM and their corresponding increased levels in the blood. In vitro stimulation with the EPCR ligand, aPC, which we found to be physiologically expressed adjacent to small murine BM blood vessels, augmented EPCRhigh HSC adhesion and further limited their intracellular NO content by increasing eNOS phosphorylation at Thr495 in BM HSC, causing reduced production of NO. Conversely, administration of the pro-coagulant protease thrombin to mice induced PAR1 mediated EPCR shedding from BM HSC, followed by CXCR4 upregulation on HSC, and PAR1-mediated CXCL12 secretion by BM stromal cells. Together, these events lead to loss of retention and rapid stem cell mobilization to the blood. Interestingly, shedding of EPCR was found to be mediated by elevation of intracellular NO content, leading to EPCR co-localization with Caveolin. Correspondingly, thrombin failed to induce EPCR shedding and mobilization in eNOS and PAR1 deficient mice. Additionally, we found that BM LTR-HSC functionally express the metalloproteinase TACE (ADAM17) on the cell membrane, and that in- vitro inhibition of TACE activity by a newly developed selective inhibitor, reduces thrombin- mediated EPCR shedding, suggesting the involvement of TACE in EPCR shedding and HSC mobilization. Moreover, EPCR shedding was also CXCR4 dependent, revealing a crosstalk between EPCR, PAR1 and CXCR4. HSPC mobilized by thrombin possessed increased long-term repopulation capability following transplantation into lethally irradiated recipient mice and re-synthesis of EPCR by donor HSC in the engrafted host BM. In addition, EPCR expression was re-induced on circulating stem cells following in vitro treatment with eNOS inhibitor. Interestingly, bypassing eNOS by directly injecting NO donor, induced EPCR shedding, CXCR4 upregulation and rapid HSPC mobilization in both wild type and eNOS KO mice. Importantly, we found that similar to mice, EPCR was selectively and highly expressed by primitive human BM CD34+CD38- HSC, but not in the blood circulation of clinical G-CSF mobilized stem cells or in motile cord blood stem cells. Human BM CD34+/CD38- HSC are functionally EPCRhigh cells, maintaining low levels of intracellular NO which mediates their increased adhesion, while EPCR shedding was important for their migration and mobilization. In the functional pre-clinical NOD/SCID mouse model, G-CSF mobilization induced EPCR shedding, up-regulation of PAR1 and CXCR4 on human stem and progenitor cells, while NO signaling inhibition blocked G-CSF induced mobilization and increased both murine and human EPCRhigh stem cell accumulation in the murine BM. Our results define functional roles for EPCR, on both human and murine HSC, and suggest that regulation of EPCR expression is linked to NO, PAR1 and CXCR4 signaling as a pivotal mechanism determining HSC localization and function. Our study reveals that activation of coagulation in the context of cell injury controls stem cells retention and motility, and suggests that targeting this system may be useful in improving clinical stem cell mobilization and transplantation protocols. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
50. Cathecholamines Differently Regulate Human AML and Normal Hematopoietic Progenitor Cell Motility Via miR126 and RGS16
- Author
-
Alexander Kalinkovich, Abraham Avigdor, Arnon Nagler, Chiara Medaglia, Menachem Bitan, Tomer Itkin, and Tsvee Lapidot
- Subjects
Myeloid ,Immunology ,Wnt signaling pathway ,CD34 ,Cell migration ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CXCR4 ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Progenitor cell ,Stem cell - Abstract
Introduction Motility, proliferation, bone marrow (BM) retention and egress to the circulation of hematopoietic stem and progenitor cell (HSPCs) are key elements in normal hematopoiesis and also in the pathogenesis of acute myeloid leukemia (AML). HSPCs are tightly regulated in the BM niche by various molecules. Among them, CXCL12 and its major receptor CXCR4, expressed by both HSPCs and BM niche components, govern HSPC retention in the BM. Administration of CXCR4 antagonists lead to mobilization of both normal HSPCs and leukemia-inducing stem cells (LICs) from the BM to the circulation. We have previously found that normal human HSPCs functionally express b2-adrenergic receptors (b2-AR) that are up-regulated by myeloid cytokines such as G-CSF. The catecholaminergic neurotransmitters epinephrine and norepinephrine (NE) activate both Wnt and GSK3β signaling pathways via b2-AR, leading to enhanced HSPC proliferation, motility and BM repopulation (Spiegel et al, Nat Immunol 2007; Lapid et al, JCI 2013). These findings indicate HSPC regulation by dynamic interactions of the sympathetic nervous and hematopoietic systems. However, the role of catecholamines in regulation of AML remains elusive. Results We found that several human AML cell lines from different FAB subtypes express b2-AR. NE, a b2-AR activating ligand, increased b2-AR expression and the CXCL12-induced migration of AML primary patients’ cells and cell lines. In addition, NE treatment significantly enhanced CXCL12induced actin polymerization, which drives most of the cellular movements. Looking for a downstream effector of b2-AR, we focused on RGS16, a G-protein signaling regulator, which negatively regulates CXCL12/CXCR4 axis (Berthebaud et al., Blood 2005). Concurrently with the increase in cell migration, NE decreased RGS16 expression (both in protein and mRNA level) in monocytic AML cells. However, no effect on either cell migration or RGS16 expression was observed in non-monocytic AML cells. These data provided evidence for the involvement of catecholamines in the regulation of AML cell migration and showed a correlation between AML FAB subtypes, cell motility and RGS16 expression. One of the regulators of RGS16 levels is miR126, which is highly expressed in AML and normal HSPCs and plays an important role in mobilization and proliferation of normal HSPCs. Indeed, in search for the mechanisms underlying the above observed differences, we found that the enhancing effect of NE on CXCL12-induced migration of monocytic AML cells was accompanied by up-regulation of miR126 expression concurrently with down-regulation of RGS16 expression, whereas in non-monocytic AML cells we observed the opposite effects, suggesting that NE differently regulates AML cells belonging to different FAB subtypes. Upon studying human normal HSPCs, we found that in steady state, normal cells express low levels of β2-AR and NE did not affect either RGS16 expression or CXCL12-induced migration of both mononuclear and CD34+ cells derived from human cord blood and BM. Conclusions Our results demonstrate that while normal and AML cells share common mechanisms that govern their motility, there are unrevealed yet mechanisms, apparently cell-type dependent, which uniquely lead to opposite effects in normal HSPCs, monocytic and non-monocytic AML cells. Altogether, these findings suggest that targeting of miR126 and RGS16 pathways by specific agonists and antagonists may serve as a new approach for selective eradication of LICs. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.