Hematopoiesis/Hematopoiesis Physiology

Similar documents
Haematopoietic stem cells

CD34+ Cells: A Comparison of Stem and Progenitor Cells in Cord Blood, Peripheral Blood, and the Bone Marrow

Hematopoiesis. - Process of generation of mature blood cells. - Daily turnover of blood cells (70 kg human)

Hematopoiesis. BHS Liège 27/1/2012. Dr Sonet Anne UCL Mont-Godinne

Getting to the root of Cancer

The Role of the Embryonic Microenvironment in Hematopoietic Cell Development

Meeting Report. From December 8 to 11, 2012 at Atlanta, GA, U.S.A

Stem cells: units of development and regeneration. Fernando D. Camargo Ph.D. Whitehead Fellow Whitehead Institute for Biomedical Research.

Modeling Developmental Hematopoiesis Using Pluripotent Stem Cells

Cytokines, adhesion molecules and apoptosis markers. A comprehensive product line for human and veterinary ELISAs

Cell signalling pathways in the HSC niche. Dr. Abdullah Aljedai

The Development of Lymphocytes: B Cell Development in the Bone Marrow & Peripheral Lymphoid Tissue Deborah A. Lebman, Ph.D.

Stem cells are undifferentiated cells which are maintained within a specific niche. A stem cell

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

Generation of post-germinal centre myeloma plasma B cell.

Tissue renewal and Repair. Nisamanee Charoenchon, PhD Department of Pathobiology, Faculty of Science

DISCOVERING ATCC IMMUNOLOGICAL CELLS - MODEL SYSTEMS TO STUDY THE IMMUNE AND CARDIOVASCULAR SYSTEMS

Ig light chain rearrangement: Rescue pathway

ONTOGENY: DEVELOPMENT OF T AND B CELLS

Hematopoiesis. Hematopoiesis. Hematopoiesis

Overview B cell development T cell development

CYTOKINE RECEPTORS AND SIGNAL TRANSDUCTION

CLINICAL USE OF CELLULAR SUBPOPULATION ANALYSIS IN BM

stem cell products Basement Membrane Matrix Products Rat Mesenchymal Stem Cell Growth and Differentiation Products

Mouse Embryonic Hematopoietic Lineage Differentiation In Vitro

Follicular Lymphoma. ced3 APOPTOSIS. *In the nematode Caenorhabditis elegans 131 of the organism's 1031 cells die during development.

Lymphoid architecture & Leukocyte recirculation. Thursday Jan 26th, 2017

Hematopoietic stem cells. Presentation outline: Embryonic und adult tissue stem cells. Hematopoietic stem cells

Flow Cytometry. What is flow cytometry?

Production of the Formed Elements *

Animal Tissue Culture SQG 3242 Biology of Cultured Cells. Dr. Siti Pauliena Mohd Bohari

Seeds and soil theory by Stephen Paget at the end of the XIX century.

4. TEXTBOOK: ABUL K. ABBAS. ANDREW H. LICHTMAN. CELLULAR AND MOLECULAR IMMUNOLOGY. 5 TH EDITION. Chapter 2. pg

Bone Marrow Stroma in Myelodysplastic Syndromes

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

Chapter 3, Part A (Pages 37-45): Leukocyte Migration into Tissues

The Role of Microenvironment in the Control of Tumor Angiogenesis

Effector T Cells and

Nature Immunology: doi: /ni.3412

T Cell Development. Xuefang Cao, MD, PhD. November 3, 2015

Production of the Formed Elements (Chapter 11) *

Adaptive immune responses: T cell-mediated immunity

Signaling Vascular Morphogenesis and Maintenance

ANAT3231: lectures overview

T Cell Effector Mechanisms I: B cell Help & DTH

CYTOKINES. Based on: Cellular and Molecular Immunology, 4 th ed.,abbas A.K., Lichtman A.H. and Pober J.S. Sounders company; Philadelphia, 2010.

Regenerative Medicine for Cardiomyocytes

Allergy and Immunology Review Corner: Chapter 1 of Immunology IV: Clinical Applications in Health and Disease, by Joseph A. Bellanti.

ANAT3231: lectures overview

CD90 + Human Dermal Stromal Cells Are Potent Inducers of FoxP3 + Regulatory T Cells

Muscle Stem Cells in Regeneration

Role of Inflammatory and Progenitor Cells in Pulmonary Vascular Remodeling: Potential Role for Targeted Therapies. Traditional Hypothesis Stress

Tissue repair. (3&4 of 4)

ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY

The first wave of B lymphopoiesis develops independent of stem cells in the murine

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

IMMU 7630 Fall 2011 ONTOGENY: DEVELOPMENT OF T AND B CELLS

Childhood hematopoiesis and hematological features. Yongmin Tang Dept. Hematology-oncology Chidlren s Hospital Zhejiang University School of medicine

Myeloproliferative Disorders - D Savage - 9 Jan 2002

Tumor Associated Macrophages as a Novel Target for Cancer Therapy

Chapter 11. B cell generation, Activation, and Differentiation. Pro-B cells. - B cells mature in the bone marrow.

Agenda. Components of blood. Blood is Fluid Connective Tissue. Blood: General functions

Oncolytic Virotherapy: Targeting Cancer Stem Cells

THE HYPOXIC HEMATOPOIETIC STEM CELL NICHE Consequences of Hypoxiainduced Transcription on Stem Cell Fate

Flow Cytometry. Hanan Jafar (2017)

Bone Cell Precursors and the Pathophysiology of Bone Loss

MACROPHAGE "MONOCYTES" SURFACE RECEPTORS

Blood & Blood Formation

COMPOSITION OF BLOOD AND NORMAL ERYTHROPOIESIS

Src-INACTIVE / Src-INACTIVE

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

BMP4/Smad5 dependent stress erythropoiesis is required for the expansion of erythroid progenitors during fetal development

Chapter 2 (pages 22 33): Cells and Tissues of the Immune System. Prepared by Kristen Dazy, MD, Scripps Clinic Medical Group

Formation of Blood Cells

Supplementary Table S1. Primers used for quantitative real-time polymerase chain reaction. Marker Sequence (5 3 ) Accession No.

TCR, MHC and coreceptors

Stem Cells. Induced Stem Cells

Chapter 11. B cell generation, Activation, and Differentiation. Pro-B cells. - B cells mature in the bone marrow.

Dynamic niches in the origination and differentiation of haematopoietic stem cells

Index. endocytosis, 92 fat cells, 99 myelofibrosis, 390 nerves, 99, 100 sinuses, 90 Bone marrow fatty involution, red and yellow marrow,

The recruitment of leukocytes and plasma proteins from the blood to sites of infection and tissue injury is called inflammation

Basis of Immunology and

Immunopathology. 2-Patterned hemodynamic responses, cell surface associated and soluble mediator systems (e.g., complement and coagulation systems).

Characterization of human myeloid progenitors and their differentiation

Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future

PBS Class #2 Introduction to the Immune System part II Suggested reading: Abbas, pgs , 27-30

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center

The Immune System. A macrophage. ! Functions of the Immune System. ! Types of Immune Responses. ! Organization of the Immune System

PhD THESIS Epigenetic mechanisms involved in stem cell differentiation

Development of the hematopoietic system in the mouse

The Adaptive Immune Response. T-cells

Recommended reading: Abbas et al. 5th edition, chapters 7 and 10; Janeway and Travers, 5th edition, chapter 7.

Hematopoie)c System. Kris)ne Kra2s, M.D.

Immune response to infection

The development of T cells in the thymus

Normal & Leukaemic haematopoiesis. Dr. Liu Te Chih Dept of Haematology / Oncology National University Health Services Singapore

Current Developments in Mobilization of Hematopoietic Stem and Progenitor Cells and Their Interaction with Niches in Bone Marrow

Accelerate Your Research with Conversant Bio

Foundations in Microbiology

Transcription:

Hematopoiesis/Hematopoiesis Physiology Definitions Hematopoiesis is the process of continuous generation of mature blood cells in the bone marrow (Figure 1). Blood cells represent different kinds of mature circulating cells, their precursors in the bone marrow or the lymphatic organs, and the progenitor and stem cells, residing in the adult marrow, but also circulating in blood in some instances. Hematopoietic stem cells are specialized, rare cells, sharing some very important basic properties, namely: Self renewal Extensive proliferative capacity Multipotent differentiation Ability to reconstitute hematopoiesis in compromised recipients Probably represent the target for leukemic transformation Quiescence Plasticity (?) These properties, except the last one, are considered essential for the definition of stem cells; plasticity, a recent hypothesis, is currently under investigation. Stem cells are extremely rare. The incidence of reconstituting stem cells is estimated at 1 to 2.5 per 100,000 injected nucleated marrow cells. Progenitor cells When get into active hematopoiesis, they exit the Go phase of the cell cycle, and undergo a series of maturational cell divisions, leading to the generation of progenitor cells. Progenitor cells are characterized by: Inability to reconstitute hematopoiesis in vivo Limited proliferative capacity Decreased or no self renewal capacity Active cycling Irreversible lineage commitment THE HIERARCHICAL MODEL OF HEMATOPOIESIS assumes that hematopoiesis involves a stepwise process where multipotent stem cells give rise to a hierarchy of progenitor cell populations. During this process proliferative potential and ability for self renewal are gradually lost, whereas differentiation characteristics are acquired (Figure 2). T-lymphocyte platelets megakaryocyte B-lymphocyte NK-cell eosinophil Hematopoietic stem cell basophil neutrophil erhythrocytes monocyte FIGURE 1: Hematopoiesis. 9

self-renewal proliferation Pre-T cell T-lymphocyte self-renewal Common lymphoid progenitor Pre-B cell NK progenitor B-lymphocyte NK-cell BFU-E erhythrocyte Hematopoietic stem cell CFU-Meg megakaryocyte plateles Common myeloid progenitor CFU-Ba CFU-Eo basophil eosinophil CFU-G neutrophil FIGURE 2: Hierarchical model of hematopoiesis. CFU-GM CFU-M monocyte commitment differentiation THE CONTINUUM MODEL OF HEMATOPOIESIS, more recently proposed, suggests that the engraftable stem cell and the progenitor cell population are the same cell population which evidences different phenotypes at different points in the cell cycle, continuously and reversibly changing its gene expression profile and, thus, its surface receptor expression (Figure 3). G 2 /M progenitor 2 G 0 /G 1 stem cell S FIGURE 3: Continuum model of hematopoiesis. progenitor 1 10

H E M A T O P O I E S I S Stemness Stemness is defined as the pattern of gene expression that is common in all stem cells. As self renewal and pluripotency are the main stem cell properties, most of the stemness genes are believed to be related to these functions. In a more extended view, stemness genes may be shared by stem cells from different tissues, although in two studies aiming to prove this, only one gene was common among the 3 purified stem cell populations tested. It is uncertain at this point whether this finding casts doubt on the stemness hypothesis or on the methods used to prove it. Despite these facts, it is already known that certain genes are expressed more often in stem cell populations, permitting investigators to propose a set of stemness genes. Negativity for cell lineage markers, proliferation and self renewal connected genes and markers permitting interaction with specific microenvironments are included. It is most certain that no single marker will emerge to be unique for stemness and at the same time we could also suppose that the absence of a significant number of these genes from a cell population will rather not make these cells fit for the title of stem cells. ARF MDM2 p53 BMI1 p16 INK4A p21 CIP1 p18 INK4C p27 KIP1 mitogenic stimuli CDK 4/6 cyclin CDK 2 D cyclin E prb E2F P P prb E2F P P P P P prb E2F Go/G1 S FIGURE 4: Cell cycle regulation of hematopoietic stem cells. Cell cycle regulation of hematopoietic stem cells Quiescence versus Proliferation or activation Most s are believed to reside in their niches in a quiescent state, at the Go phase of the cell cycle. This state is better described as a state of readiness, as s are able to respond to appropriate signals and proliferate. Various gene products have been implicated in this process of stem cell arousal, which is characterized by a phase of preparation and subsequent phases of early and late proliferation. A re induction of quiescence marks the final stage of activation cyscle. Cell cycle is regulated in s via the actions of specific gene products on cyclin cyclin dependent kinases (cdk) complexes. Of importance among these cell cycle regulating genes seem to be Bmi 1, p21, p27, p18 and arf (Figure 4). Recent experiments have shown that the initial phase of exit from quiescence are mostly stochastic or intrinsic to the, while later phases are increasingly induced by signals from the microenvironment of the hematopoietic niche. As it has been recently reported, constitutive activation of NF κb is not sufficient to disturb normal steady state hematopoiesis. 11

ECM integrins Wnt/frizzled Notch/jagged telomerase stroma cell transcription factors self-renewal HOXB 4 cell cycle regulators ILs and other soluble GFs differentiation apoptosis mobilization FIGURE 5: Molecular basis of hematopoietic stem cell fate decisions. Hematopoietic stem cell fate decisions Self renewal versus Differentiation versus Apoptosis Commitment to terminal differentiation and self renewal represent two opposite outcomes. Normal hematopoiesis requires a balance between these two outcomes. Two models have been proposed to explain the mechanisms that regulate the decision for self renewal or differentiation: the stochastic and the deterministic model. In the stochastic model the decision of an individual stem cell to undergo self renewal or differentiation is thought to be determined by chance. In the deterministic model, the stem cell fate is determined by the action of cytokines and extracellular matrix components (Figure 5). As already stated most stem cells are in a state of quiescence. They, however, do divide and during these divisions some crucial decision is taken between three mainly fates, namely self renewal, differentiation or apoptosis. Intrinsic and extrinsic factors from the BM microenvironment regulate the fate. The non hematopoietic cells from the BM microenvironment can interact with the hematopoietic progenitors through integrins, adhesion molecules, and receptor ligand juxtaposition (i.e. Delta/Notch1, ckit/scf). Additionally these cells can secrete various factors (i.e. chemokines and growth factors) and they can synthesize and display on their cell surface various forms of proteoglycans to enhance cell cell interactions and sequester soluble factors within the BM to. A number of gene products have been implicated in the self renewal versus differentiation process of s. Among these genes Wnt, Notch and HoxB4 seem to play especially important roles. Wnt promotes nuclear translocation of β catenin resulting in increased self renewal of. Notch maintains the pluripotent identity of. The prevailing opinion is that expression of the above genes drives stem cells towards self renewal and preservation of stemness, instead of differentiation. Absence of their collaborative expression, on the other hand, leads as a default choice towards differentiation. Self renewal implies conservation of s numbers and this seems to be mediated via asymmetric divisions. This means that in each division one of the daughter cells maintains stemness and the other is marked for differentiation. Little is understood concerning the basis of this distinction and the signaling pathways involved, although recently substantial progress is made, mostly from studies in lower organisms, where polarity of s is a crucial factor. An important role for the Smad signaling pathway in the regulation of self renewal of s has been reported in an model. Apoptosis is another probable fate of s and its role in normal hematopoiesis is strongly debated. A role for apoptosis and its defects in cancer cells is better understood. 12

H E M A T O P O I E S I S Marrow microenvironment Marrow microenvironment is a term used to denote the specialized milieu of the bone marrow cavity, in which hematopoiesis takes place in the adult (Figure 6). Niche is defined as the specific in vivo regulatory microenvironment where s reside. This structure is composed from different cell types, which contribute via cell receptors and soluble factors to the localization, survival, self renewal and differentiation of s (Figure 7 and 8). This niche concept includes a developmental aspect as well, as different aggregations of cells are supporting hematopoiesis in the AGM region of the embryo, the fetal liver or the marrow. niche bone osteoblasts ECM myofibroblast Endothelial cells Marrow fibroblasts, myofibroblasts and endothelial cells are considered components of the hematopoietic inductive environment; recently the role of endosteal osteoblasts and their precursors has gained considerable recognition, as a prominent player in the process. fibroblast The number of niches is probably determined by PTH stimulated osteoblastic proliferation. Homing of s is their ability to localize and reside at the specialized niches, where hematopoiesis can take place. This is achieved by a dynamic process involving mutual recognition and continuous interaction with stromal cells, via a complex network of surface molecules. BMP FIGURE 6: Bone marrow microenvironment. PTH Mobilization of s may act as a regulator of the hematopoietic stem cell compartment size. Mobilization could be a death pathway, a mechanism that regulates stem cell number. If a replication event occurs and a niche is not available, one offspring mobilizes/dies. After cytokine exposure, many exit the marrow, travel through blood and then repopulate empty marrow niches. FIGURE 7: Regulation of homing, survival, and maintenance by the niche. Endosteal osteoblasts are prominent components of the niche. Osteoblast interactions through cell cell adhesion molecules (N cadherin, β1 integrins, VLA 4 VCAM, LFA ICAM etc), soluble and cell surface associated molecules (Ang 1, Jagged 1, osteopontin), cytokines and growth factors (G CSF, GM CSF, M CSF, IL 1, SDF 1, TGF β etc) regulate stem cell niche localization, survival, quiesence or proliferation and play a key role in the establishment and maintenance of the niche in the bone marrow (BM). Homophilic interaction through N cadherin apears to be critical in keeping s quiescent and may also provide a link to Wnt B catenin signaling pathway that has been shown to regulate self renewal. niche localization and self renewal in the osteoblastic niche is also critically regulated by Jagged 1 (expressed on osteoblasts) Notch signaling. Osteoblasts also secrete Ang 1 which, by activating of Tie2 receptors on s, promotes tight adhesion of stem cells in their niche and stem cell maintenance. osteopontin Cytokines GFs B-integrin osteoblast ICAM-LFA VCAM-VLA ECM N-cadherin B-catenin Homing Quiescence Survival Jagged Notch Tie2 Ang1 M c-kit BMSC These interactions are likely influenced by signals such as PTH and BMPs that regulate osteoblastic function. However, despite the prominent role of osteoblasts, other bone marrow stromal cells (BMSCs) as well as attachment to extracellular matrix (ECM) are also requiered for stem cell maintenance e.g the quiescent is kept in the osteoblastic niche in cooperation with adjacent membrane bound c kit expressing BMSCs. : haematopoietic stem cell, BMPs: Bone morphogenetic proteins, PTH: parathormone, ICAM: intracellular cell adhesion molecule, LFA: lymphocyte function associated antigen, VCAM: vascular cell adhesion molecule, Ang 1: angiopoietin 1, GFs: growth factors, ECM: extracellular matrix, BMSCs: bone marrow stromal cells, M c Kit: membrane bound c kit. 13

Osteoblastic niche 1 Endothelial cells Osteoblast MMP9 action S c-kit M c-kit BMSC quiescence Vascular niche proliferation 2 Osteoblast differentiation differentiation BMSC Osteoblasts back to osteoblastic niche FIGURE 8: Osteoblastic and vascular niche. Although most s reside in Go within their niches there is always a small number of s that enters the cell cycle to initiate self renewal or differentiation. It is not clear whether proliferation and differentiation occurs into the osteoblastic niche or involves local mobilization to osteogenic stromal cells expressing Jag 1 or vascular niches specialized for proliferation. In one proposed model the activity of matrix metalloproteinase 9 (MMP 9) expressed within the osteoblastic zone results in cleavage of the membrane kit ligant from BM stromal cells (BMSCs). Soluble kit ligant then promotes cell cycle entry and localization of stem cells in the vascular zone where proliferation and differentiation occurs (1). Alternatively assymetric division could occur in the osteoblastic zone and s daugther cells released may return to accessible osteoblastic niches or may be mobilized to the periphery or induced to differentiate (2). Ontogeny of hematopoiesis The developmental origin of adult type hematopoiesis is currently placed at the intrabody portion of the embryo, and especially the aorta gonads mesonephros (AGM) region, rather at the extra embryonic yolk sac, although both sites are rather contributing. Fetal liver is the predominant site at approximately 6 weeks of gestation until about midterm, when bone marrow replaces fetal liver (Figure 9). In some species, as the mouse, spleen is an adult site of hematopoiesis. Primitive and definitive hematopoietic cells in the mouse embryo arise from embryonic endothelial cell marker positive cells. An important difference between fetal and adult hematopoiesis is the inability of the marrow cavity of the fetus to expand in situations of hematopoietic stress. SCL (Stem Cell Ligand)acts not only at the level of erythroid commitment, but also at both multipotent and myeloid committed stages of adult hematopoiesis. Among the molecular mechanisms implicated in early hematopoiesis significant role has been attributed to vascular endothelial growth factor (VEGF) and its receptor flk 1, as well as to bone morphogenetic protein 4 (BMP 4), fibroblast growth factor, and hedgehog proteins. It is of importance that hematopoietic and vascular cells are thought to arise from a common progenitor called the haemangioblast, which is found at highest frequency in the posterior region of the primitive streak, indicating that initial stages of haematopoietic and vascular commitment occur before blood island development in the yolk sac. 14

H E M A T O P O I E S I S Yolk sac AGM 6 weeks liver Midtrimester Bone Marrow Spleen FIGURE 9: Ontogeny of hematopoiesis. Selected references: Arai, F., A. Hirao, et al. (2004). Tie2/angiopoietin 1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 118(2): 149 61 Beslu, N., J. Krosl, et al. (2004). Molecular interactions involved in HOXB4 induced activation of self renewal. Blood 104(8): 2307 2314 Blank, U., G. Karlsson, et al. (2006). Smad7 promotes self renewal of hematopoietic stem cells. Blood 108(13): 4246 54 de la Grange, P. B., F. Armstrong, et al. (2006). Low SCL/TAL1 expression reveals its major role in adult hematopoietic myeloid progenitors and stem cells. Blood 108(9): 2998 3004 Duncan, A. W., F. M. Rattis, et al. (2005). Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nat Immunol 6(3): 314 Ema, M., T. Yokomizo, et al. (2006). Primitive erythropoiesis from mesodermal precursors expressing VE cadherin, PECAM 1, Tie2, endoglin, and CD34 in the mouse embryo. Blood 108(13): 4018 24 Faubert, A., J. Lessard, et al. (2004). Are genetic determinants of asymmetric stem cell division active in hematopoietic stem cells? Oncogene 23(43): 7247 55 Fortunel, N. O., H. H. Otu, et al. (2003). Comment on Stemness : transcriptional profiling of embryonic and adult stem cells and a stem cell molecular signature. Science 302 (5644): 393; author reply 393 15

Ivanova, N. B., J. T. Dimos, et al. (2002). A stem cell molecular signature. Science 298(5593): 601 4 Iwama, A., H. Oguro, et al. (2004). Enhanced self renewal of hematopoietic stem cells mediated by the polycomb gene product Bmi 1. Immunity 21(6): 843 51 Lessard, J., A. Faubert, et al. (2004). Genetic programs regulating specification, maintenance and expansion. Oncogene 23(43): 7199 209 Pyle, A. D., P.J. Donovan, et al. (2004). Chipping away at 'stemness.genome Biol 5(8): 235 Quesenberry, P. J., G. A. Colvin, et al. (2002). The chiaroscuro stem cell: a unified stem cell theory. Blood 100(13): 4266 71 Ramalho Santos, M., S. Yoon, et al. (2002). Stemness: transcriptional profiling of embryonic and adult stem cells. Science 298(5593): 597 600 Reya, T., A. W. Duncan, et al. (2003). A role for Wnt signalling in self renewal of haematopoietic stem cells. Nature 423(6938): 409 Schepers, H., B. J. Eggen, et al. (2006). Constitutive activation of NF kappab is not sufficient to disturb normal steady state hematopoiesis. Haematologica 91(12): 1710 1 Srour, E. F., X. Tong, et al. (2005). Modulation of in vitro proliferation kinetics and primitive hematopoietic potential of individual human CD34+CD38 /lo cells in G0. Blood 105(8): 3109 3116 Steinman, R. A. (2002). Cell cycle regulators and hematopoiesis. Oncogene 21(21): 3403 13 Taichman, R. S. (2005). Blood and bone: two tissues whose fates are intertwined to create the hematopoietic stem cell niche. Blood 105(7): 2631 9 Venezia, T. A., A. A. Merchant, et al. (2004). Molecular Signatures of Proliferation and Quiescence in Hematopoietic Stem Cells. PLoS Biology 2(10): e301 Wagner, W., A. Ansorge, et al. (2004). Molecular evidence for stem cell function of the slow dividing fraction among human hematopoietic progenitor cells by genome wide analysis. Blood 104(3): 675 686 Wallenfang, M. R. and E. Matunis (2003). Developmental biology. Orienting stem cells. Science 301(5639): 1490 1 Zhu, J. and S. G. Emerson (2004). A new bone to pick: osteoblasts and the haematopoietic stem cell niche. Bioessays 26(6): 595 9 16