Minireview Open Access
Copyright ©2013 Baishideng. All rights reserved.
World J Hematol. Feb 6, 2013; 2(1): 1-5
Published online Feb 6, 2013. doi: 10.5315/wjh.v2.i1.1
Bone marrow cell death and proliferation: Controlling mechanisms in normal and leukemic state
Morito Kurata, Shiho Suzuki, Shinya Abe, Iichiroh Onishi, Masanobu Kitagawa, Department of Comprehensive Pathology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
Author contributions: Suzuki S, Abe S and Onishi I offered suggestions regarding the conduct of the manuscript; Kurata M and Kitagawa M wrote the manuscript.
Correspondence to: Masanobu Kitagawa, MD, PhD, Professor, Chair, Department of Comprehensive Pathology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan. masa.pth2@tmd.ac.jp
Telephone: +81-3-58035173 Fax: +81-3-58030123
Received: June 22, 2012
Revised: November 30, 2012
Accepted: January 5, 2013
Published online: February 6, 2013

Abstract

Bone marrow cell death and proliferation are regulated by multiple factors including genetic and epigenetic alterations of hematopoietic cells, crosstalk of hematopoietic cells with bone marrow mesenchymal cells through direct cell-cell interaction or cytokine/chemokine production, vascularity of the bone marrow, and interactions of sympathetic nerve system with hematopoiesis. Cell proliferation usually predominates over cell death in neoplastic processes such as leukemia and myeloproliferative neoplasms, while apoptotic processes also have a significant role in the pathogenesis of myelodysplastic syndromes. Recently, hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs) have been identified and their characters on self renewal process, differentiation, cell dynamics and drug resistance have been implicated. Although most leukemia cells are initially sensitive to chemo- or radiotherapy, LSCs are resistant and considered to be the basis for disease relapse after initial response. HSCs and LSCs may use similar interactions with bone marrow microenvironment. However, bone marrow microenvironment called niche should influence the normal as well as malignant hematopoiesis in different manners. Recent studies have expanded the number of cell types constituting bone marrow niche and made the issue more complex. Since the majority of excellent and contributing studies on bone marrow niches have been performed in animal models, niches in human tissues are beginning to be localized and characterized. In this article, we summarize the relation of hematopoietic cells with niches and hope to point a hint to the novel strategy for treatment of malignant proliferation of hematopoietic cells.

Key Words: Bone marrow, Hematopoietic stem cells, Niche, Apoptosis, Leukemia



INTRODUCTION

Cell dynamics of hematopoietic cells/leukemic cells may be regulated by multiple factors in the bone marrow. Recently, interactions of hematopoietic stem cells (HSCs) with the bone marrow microenvironment have been extensively studied mainly using animal models with genetic modification. Many signaling pathways such as CXCR4/CXCL12, Jagged/Notch, Wnt, Ang-1/Tie2, SCF, very late antigen-4 (VLA-4), VCAM-1, and TGF have been shown to regulate the self-renewal, differentiation, proliferation, and apoptosis/senescence of HSCs[1-11]. Mechanisms of crosstalk between hematopoietic cells and the bone marrow microenvironment would be closely associated with the pathogenesis of hematologic diseases including malignancies. Although leukemia cells are thought to harbor cell-autonomous mutations, the interaction with the microenvironment should play an important role in the regulation of leukemia cell dynamics[12]. This article reiterates the importance of mechanisms affecting the functions of the bone marrow microenvironment on the proliferation/apoptosis of hematopoietic/leukemia cells.

HSCS AND THEIR NICHE

The HSCs have two opposing characters - maintenance of an undifferentiated state analogous to pluripotent stem cells, and the execution of tissue-specific hematopoietic functions[13]. In the bone marrow, characters of HSCs are maintained within a special microenvironment called a niche. The concept of niches was proposed in the 1970s; however, the nature and function of these stromal structures remains unclear. HSCs are in contact with bone-lining osteoblastic cells, but only a part of the HSCs seem to be located in this endosteal niche region. Instead, many HSCs are found in association with the sinusoidal endothelium, referred to as the vascular niche. However, osteoblast depletion results in extramedullary hematopoiesis, suggesting that a vascular niche alone is not sufficient to maintain hematopoiesis[14]. Endosteal and vascular niches might regulate different HSC populations, although recent data depicts a more complicated feature, with functional crosstalk between cells in these two regions[2,15,16].

Recently it was suggested that primitive mesenchymal cells, including CXCL12-abundant reticular cells[17-20], nestin-expressing cells with sympathetic nerve system[21-23], nonmyelinating Schwann cells[24] and macrophages[25,26] act as bone marrow niche-associated cells. The interactions between CXCR4 and CXCL12 (SDF-1) are especially important in the localization and retention of HSCs in the bone marrow. Furthermore, chemokine interactions through CXCL12 can cause the up-regulation of vascular cell adhesion molecule-1 and VLA-4 expression[27]. CXCL12 also has an essential role in colonization of the bone marrow by HSCs during early development, because CXCL12-deficient embryos have severely reduced HSC numbers and a disturbed function[28]. Further studies on the interplay of such regulatory forces as “cell fate and localization determinant” will likely shed light on the pathogenesis of hematological diseases.

BONE MARROW NICHES IN HEMATOPOIETIC PATHOLOGY

Recent animal model studies have provided insights into the role of aberrant microenvironment signaling leading to the pathogenesis of hematological diseases. Perturbations in niche signaling in murine models[29] can mimic idiopathic myelofibrosis, leading to enhanced stem cell mobilization and the creation of alternate niches[30]. The deletion of the Dicer1 gene specifically in mouse osteoblasts (endosteal niches) disrupted the integrity of hematopoiesis. Myelodysplastic syndromes (MDS)-like phenotypes emerged in spite of the fact that hematopoietic cells had an intact Dicer1[31]. Therefore, perturbation of specific mesenchymal stromal cells can cause disorder in the differentiation, proliferation, and apoptosis of hematopoietic cells. In this mouse model, acute myeloid leukemia (AML) emerged with several acquired genetic abnormalities, supporting the concept of niche-induced leukemogenesis. Also, in human samples, the impaired expression of DICER-associated genes was demonstrated in mesenchymal stromal cells from MDS patients[32].

Concerning the bone marrow cell dynamics in MDS, excessive apoptosis of hematopoietic cells was observed to be induced by the bone marrow microenvironment[33-48]. The apoptosis was mediated by paracrine as well as autocrine factors, implicating both medullary stromal cells and hematopoietic cells in the pathology of the disease. Pro-inflammatory cytokines such as tumor necrosis factor in the bone marrow microenvironment are mainly paracrine mediators of apoptosis. As autocrine stimulation mechanisms, it has recently been shown that the deregulation of ribosome biogenesis can initiate a stress response in hematopoietic cells through the p53-mediated signaling pathway. Thus, both the stromal cells of the bone marrow microenvironment and hematopoietic cells themselves possess a common and characteristic biology in this heterogeneous disease entity.

In human samples, the microenvironment has also been studied in multiple myeloma[49]. Self-renewal pathway activation in the niche (such as the canonical Wnt pathway) has been postulated to result in enhanced myeloma cell survival[50]. Another example of microenvironment-associated disease is the WHIM (Warts, Hypogammaglobulinemia, Infections, and Myelokathexis) syndrome. This rare syndrome is known to exhibit congenital neutropenia often caused by mutations in CXCR4 with increased sensitivity to CXCL12[51].

LEUKEMIA STEM CELLS AND NICHES

Like many cancers, AML has been identified as a cell autonomous disorder. Namely, the genetic events leading to transformation of the normal hematopoietic cell are found within leukemia cells and are necessary and sufficient for the generation of leukemia. However, many studies have demonstrated evidence of functional heterogeneity among AML cells. In particular, there seems to be a subpopulation of AML cells referred to as “leukemia stem cells” (LSCs) that alone have long-term repopulating potential and the ability to propagate and maintain the AML phenotype[52]. To understand the difference of LSCs from HSCs, clarification of the crosstalk between LSCs and niches would be very important. Niche-mediated mechanisms promote the engraftment and survival of LSCs. The biology of LSCs has been determined by studies of primary human AML cells transplanted into NOD-SCID or NOD-SCID IL2Rγnull mice[53,54]. In these xenograft models, AML cells have a phenotypic hierarchy, which parallels that of normal hematopoietic stem/progenitor cells[1,55-58]. For example, the CD34+CD38- AML population could engraft efficiently in NOD-SCID mice, but the more differentiated CD34+CD38+ and CD34- AML cells were unable to engraft and yield colony-forming progenitors. Human AML cells preferentially engrafted in the bone marrow endosteal region in NOD-SCID IL2Rnull mice, and remained adjacent to bone marrow osteoblasts for up to 4 mo following transplantation[54]. Homing to the microenvironment appears important in sustaining the survival of LSCs. Moreover, these cells were highly enriched for quiescent cells and were resistant to cytosine arabinoside chemotherapy[59].

CROSSTALK OF LSCS WITH NICHES

Next, the specific mechanisms of niches regulating LSC fate should be clarified[60]. For example, the interaction between CXCR4 and CXCL12 is known to regulate normal HSC proliferation and survival[61-63]. What is the case in LSCs Actually, CXCR4 is also expressed on primary leukemic cells, and high-level expression of CXCR4 on AML cells is a negative prognostic factor of relapse-free and overall survival[64,65]. Treatment with the CXCR4 antagonist was shown to inhibit stromal cell-induced proliferative signals in AML cells and to increase the sensitivity to chemotherapy, resulting in the extended survival of AML-transplanted mice compared with treatment with chemotherapy alone[66]. These data suggest that LSCs possess a CXCR4-dependent homing capacity that is analogous to normal hematopoietic cells, and antagonism of the CXCR4/CXCL12 axis is a logical therapeutic strategy for the treatment of acute leukemia.

However LSCs are rather heterogeneous. Even in LSCs, different patterns of interactions with niches were identified[67]. The homing of pre-LSCs was similar to long-term HSCs, while the homing of established LSCs was most similar to that of committed myeloid progenitor and distinct from HSCs.

After homing to the bone marrow niches, leukemia cells are retained in these niches through cellular adhesion molecules such as VLA-4 and LFA-1 integrins[68]. Increased VLA-4 expression has been demonstrated to correlate with increased bone marrow blast counts in AML[69]. Furthermore, VLA-4-highly expressing AML cells show relative resistance to chemotherapy-induced apoptosis, and the administration of VLA-4 neutralizing antibodies inhibits this resistance[70].

On the other hand, the leukemia cells were able to directly modulate the niche at the expense of normal hematopoietic stem and progenitor cells by down-regulating CXCL12 levels in areas of leukemia infiltration[71]. It has been shown that stem cell factor, a niche regulator, was secreted by the leukemia cells, leading to the abnormal retention and engraftment of normal HSCs in the microenvironment with leukemic infiltration. Thus, identifying the mechanisms involved in the generation of various signals by HSCs, LSCs, and niches might provide new insights into the pathogenesis of acute leukemia as well as MDS and myeloproliferative neoplasms.

Footnotes

P- Reviewers Bonnet D, Fukuda S, Poggi A S- Editor Song XX L- Editor A E- Editor Zheng XM

References
1.  Lane SW, Scadden DT, Gilliland DG. The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood. 2009;114:1150-1157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 368]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
2.  Lilly AJ, Johnson WE, Bunce CM. The haematopoietic stem cell niche: new insights into the mechanisms regulating haematopoietic stem cell behaviour. Stem Cells Int. 2011;2011:274564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
3.  Oh IH, Humphries RK. Concise review: Multidimensional regulation of the hematopoietic stem cell state. Stem Cells. 2012;30:82-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
4.  Doan PL, Chute JP. The vascular niche: home for normal and malignant hematopoietic stem cells. Leukemia. 2012;26:54-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 99]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
5.  Nagasawa T, Hirota S, Tachibana K, Takakura N, Nishikawa S, Kitamura Y, Yoshida N, Kikutani H, Kishimoto T. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996;382:635-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1845]  [Cited by in F6Publishing: 1743]  [Article Influence: 62.3]  [Reference Citation Analysis (0)]
6.  Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature. 1998;393:595-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1914]  [Cited by in F6Publishing: 1850]  [Article Influence: 71.2]  [Reference Citation Analysis (0)]
7.  Batard P, Monier MN, Fortunel N, Ducos K, Sansilvestri-Morel P, Phan T, Hatzfeld A, Hatzfeld JA. TGF-(beta)1 maintains hematopoietic immaturity by a reversible negative control of cell cycle and induces CD34 antigen up-modulation. J Cell Sci. 2000;113:383-390.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, Ito K, Koh GY, Suda T. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118:149-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1457]  [Cited by in F6Publishing: 1408]  [Article Influence: 70.4]  [Reference Citation Analysis (0)]
9.  Mancini SJ, Mantei N, Dumortier A, Suter U, MacDonald HR, Radtke F. Jagged1-dependent Notch signaling is dispensable for hematopoietic stem cell self-renewal and differentiation. Blood. 2005;105:2340-2342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 204]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
10.  Maillard I, Koch U, Dumortier A, Shestova O, Xu L, Sai H, Pross SE, Aster JC, Bhandoola A, Radtke F. Canonical notch signaling is dispensable for the maintenance of adult hematopoietic stem cells. Cell Stem Cell. 2008;2:356-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 248]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
11.  Butler JM, Nolan DJ, Vertes EL, Varnum-Finney B, Kobayashi H, Hooper AT, Seandel M, Shido K, White IA, Kobayashi M. Endothelial cells are essential for the self-renewal and repopulation of Notch-dependent hematopoietic stem cells. Cell Stem Cell. 2010;6:251-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 488]  [Cited by in F6Publishing: 486]  [Article Influence: 34.7]  [Reference Citation Analysis (0)]
12.  Vas V, Wandhoff C, Dörr K, Niebel A, Geiger H. Contribution of an aged microenvironment to aging-associated myeloproliferative disease. PLoS One. 2012;7:e31523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
13.  Forsberg EC, Passegué E, Prohaska SS, Wagers AJ, Koeva M, Stuart JM, Weissman IL. Molecular signatures of quiescent, mobilized and leukemia-initiating hematopoietic stem cells. PLoS One. 2010;5:e8785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 104]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
14.  Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood. 2004;103:3258-3264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 571]  [Cited by in F6Publishing: 534]  [Article Influence: 26.7]  [Reference Citation Analysis (0)]
15.  Lo Celso C, Fleming HE, Wu JW, Zhao CX, Miake-Lye S, Fujisaki J, Côté D, Rowe DW, Lin CP, Scadden DT. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009;457:92-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 672]  [Cited by in F6Publishing: 657]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
16.  Ehninger A, Trumpp A. The bone marrow stem cell niche grows up: mesenchymal stem cells and macrophages move in. J Exp Med. 2011;208:421-428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in F6Publishing: 399]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
17.  Nagasawa T, Omatsu Y, Sugiyama T. Control of hematopoietic stem cells by the bone marrow stromal niche: the role of reticular cells. Trends Immunol. 2011;32:315-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 91]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
18.  Kiel MJ, Morrison SJ. Maintaining hematopoietic stem cells in the vascular niche. Immunity. 2006;25:862-864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 103]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
19.  Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25:977-988.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1639]  [Cited by in F6Publishing: 1660]  [Article Influence: 97.6]  [Reference Citation Analysis (0)]
20.  Sugiyama T, Nagasawa T. Bone marrow niches for hematopoietic stem cells and immune cells. Inflamm Allergy Drug Targets. 2012;11:201-206.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Méndez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, Scadden DT, Ma’ayan A, Enikolopov GN, Frenette PS. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2398]  [Cited by in F6Publishing: 2439]  [Article Influence: 174.2]  [Reference Citation Analysis (0)]
22.  Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, Frenette PS. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 993]  [Cited by in F6Publishing: 971]  [Article Influence: 53.9]  [Reference Citation Analysis (0)]
23.  Méndez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 921]  [Cited by in F6Publishing: 915]  [Article Influence: 57.2]  [Reference Citation Analysis (0)]
24.  Yamazaki S, Ema H, Karlsson G, Yamaguchi T, Miyoshi H, Shioda S, Taketo MM, Karlsson S, Iwama A, Nakauchi H. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell. 2011;147:1146-1158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 540]  [Cited by in F6Publishing: 545]  [Article Influence: 45.4]  [Reference Citation Analysis (0)]
25.  Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, Poulton IJ, van Rooijen N, Alexander KA, Raggatt LJ. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116:4815-4828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 584]  [Cited by in F6Publishing: 583]  [Article Influence: 41.6]  [Reference Citation Analysis (0)]
26.  Chow A, Lucas D, Hidalgo A, Méndez-Ferrer S, Hashimoto D, Scheiermann C, Battista M, Leboeuf M, Prophete C, van Rooijen N. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011;208:261-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 591]  [Cited by in F6Publishing: 600]  [Article Influence: 46.2]  [Reference Citation Analysis (0)]
27.  Avecilla ST, Hattori K, Heissig B, Tejada R, Liao F, Shido K, Jin DK, Dias S, Zhang F, Hartman TE. Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med. 2004;10:64-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 559]  [Article Influence: 26.6]  [Reference Citation Analysis (0)]
28.  Ara T, Tokoyoda K, Sugiyama T, Egawa T, Kawabata K, Nagasawa T. Long-term hematopoietic stem cells require stromal cell-derived factor-1 for colonizing bone marrow during ontogeny. Immunity. 2003;19:257-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 271]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
29.  Walkley CR, Olsen GH, Dworkin S, Fabb SA, Swann J, McArthur GA, Westmoreland SV, Chambon P, Scadden DT, Purton LE. A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency. Cell. 2007;129:1097-1110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 426]  [Cited by in F6Publishing: 399]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
30.  Lataillade JJ, Pierre-Louis O, Hasselbalch HC, Uzan G, Jasmin C, Martyré MC, Le Bousse-Kerdilès MC. Does primary myelofibrosis involve a defective stem cell niche From concept to evidence. Blood. 2008;112:3026-3035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 106]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
31.  Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, Ebert BL, Al-Shahrour F, Hasserjian RP, Scadden EO. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464:852-857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 797]  [Cited by in F6Publishing: 811]  [Article Influence: 57.9]  [Reference Citation Analysis (0)]
32.  Santamaría C, Muntión S, Rosón B, Blanco B, López-Villar O, Carrancio S, Sánchez-Guijo FM, Díez-Campelo M, Alvarez-Fernández S, Sarasquete ME. Impaired expression of DICER, DROSHA, SBDS and some microRNAs in mesenchymal stromal cells from myelodysplastic syndrome patients. Haematologica. 2012;97:1218-1224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
33.  Kitagawa M, Kurata M, Yamamoto K, Abe S, Suzuki S, Umeda S. Molecular pathology of myelodysplastic syndromes: biology of medullary stromal and hematopoietic cells (review). Mol Med Rep. 2011;4:591-596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
34.  Kitagawa M, Kamiyama R, Kasuga T. Expression of the proliferating cell nuclear antigen in bone marrow cells from patients with myelodysplastic syndromes and aplastic anemia. Hum Pathol. 1993;24:359-363.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Kitagawa M, Yoshida S, Kuwata T, Tanizawa T, Kamiyama R. p53 expression in myeloid cells of myelodysplastic syndromes. Association with evolution of overt leukemia. Am J Pathol. 1994;145:338-344.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Kitagawa M, Saito I, Kuwata T, Yoshida S, Yamaguchi S, Takahashi M, Tanizawa T, Kamiyama R, Hirokawa K. Overexpression of tumor necrosis factor (TNF)-alpha and interferon (IFN)-gamma by bone marrow cells from patients with myelodysplastic syndromes. Leukemia. 1997;11:2049-2054.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Kitagawa M, Yamaguchi S, Takahashi M, Tanizawa T, Hirokawa K, Kamiyama R. Localization of Fas and Fas ligand in bone marrow cells demonstrating myelodysplasia. Leukemia. 1998;12:486-492.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Kitagawa M, Takahashi M, Yamaguchi S, Inoue M, Ogawa S, Hirokawa K, Kamiyama R. Expression of inducible nitric oxide synthase (NOS) in bone marrow cells of myelodysplastic syndromes. Leukemia. 1999;13:699-703.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Mundle SD, Ali A, Cartlidge JD, Reza S, Alvi S, Showel MM, Mativi BY, Shetty VT, Venugopal P, Gregory SA. Evidence for involvement of tumor necrosis factor-alpha in apoptotic death of bone marrow cells in myelodysplastic syndromes. Am J Hematol. 1999;60:36-47.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Mundle SD, Reza S, Ali A, Mativi Y, Shetty V, Venugopal P, Gregory SA, Raza A. Correlation of tumor necrosis factor alpha (TNF alpha) with high Caspase 3-like activity in myelodysplastic syndromes. Cancer Lett. 1999;140:201-207.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Allampallam K, Shetty V, Mundle S, Dutt D, Kravitz H, Reddy PL, Alvi S, Galili N, Saberwal GS, Anthwal S. Biological significance of proliferation, apoptosis, cytokines, and monocyte/macrophage cells in bone marrow biopsies of 145 patients with myelodysplastic syndrome. Int J Hematol. 2002;75:289-297.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Sawanobori M, Yamaguchi S, Hasegawa M, Inoue M, Suzuki K, Kamiyama R, Hirokawa K, Kitagawa M. Expression of TNF receptors and related signaling molecules in the bone marrow from patients with myelodysplastic syndromes. Leuk Res. 2003;27:583-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 87]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
43.  Yamamoto K, Abe S, Nakagawa Y, Suzuki K, Hasegawa M, Inoue M, Kurata M, Hirokawa K, Kitagawa M. Expression of IAP family proteins in myelodysplastic syndromes transforming to overt leukemia. Leuk Res. 2004;28:1203-1211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 61]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
44.  Kurata M, Hasegawa M, Nakagawa Y, Abe S, Yamamoto K, Suzuki K, Kitagawa M. Expression dynamics of drug resistance genes, multidrug resistance 1 (MDR1) and lung resistance protein (LRP) during the evolution of overt leukemia in myelodysplastic syndromes. Exp Mol Pathol. 2006;81:249-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
45.  Bourne JR, Jagannathan V, Giese B, Ward JW. A software system for syntactic analysis of the EEG. Comput Programs Biomed. 1980;11:190-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
46.  Kuninaka N, Kurata M, Yamamoto K, Suzuki S, Umeda S, Kirimura S, Arai A, Nakagawa Y, Suzuki K, Kitagawa M. Expression of Toll-like receptor 9 in bone marrow cells of myelodysplastic syndromes is down-regulated during transformation to overt leukemia. Exp Mol Pathol. 2010;88:293-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
47.  Raza A, Cruz R, Latif T, Mukherjee S, Galili N. The biology of myelodysplastic syndromes: unity despite heterogeneity. Hematol Rep. 2010;2:e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
48.  Suzuki S, Kurata M, Abe S, Miyazawa R, Murayama T, Hidaka M, Yamamoto K, Kitagawa M. Overexpression of MCM2 in myelodysplastic syndromes: association with bone marrow cell apoptosis and peripheral cytopenia. Exp Mol Pathol. 2012;92:160-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
49.  Podar K, Chauhan D, Anderson KC. Bone marrow microenvironment and the identification of new targets for myeloma therapy. Leukemia. 2009;23:10-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 255]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
50.  Derksen PW, Tjin E, Meijer HP, Klok MD, MacGillavry HD, van Oers MH, Lokhorst HM, Bloem AC, Clevers H, Nusse R. Illegitimate WNT signaling promotes proliferation of multiple myeloma cells. Proc Natl Acad Sci USA. 2004;101:6122-6127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 250]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
51.  Kawai T, Choi U, Cardwell L, DeRavin SS, Naumann N, Whiting-Theobald NL, Linton GF, Moon J, Murphy PM, Malech HL. WHIM syndrome myelokathexis reproduced in the NOD/SCID mouse xenotransplant model engrafted with healthy human stem cells transduced with C-terminus-truncated CXCR4. Blood. 2007;109:78-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 67]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
52.  Kikushige Y, Shima T, Takayanagi S, Urata S, Miyamoto T, Iwasaki H, Takenaka K, Teshima T, Tanaka T, Inagaki Y. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell. 2010;7:708-717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 337]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
53.  Somervaille TC, Cleary ML. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell. 2006;10:257-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 438]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
54.  Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, Nakamura R, Tanaka T, Tomiyama H, Saito N. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 2007;25:1315-1321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 705]  [Cited by in F6Publishing: 712]  [Article Influence: 41.9]  [Reference Citation Analysis (0)]
55.  Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3316]  [Cited by in F6Publishing: 3222]  [Article Influence: 107.4]  [Reference Citation Analysis (0)]
56.  Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat Immunol. 2004;5:738-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 691]  [Cited by in F6Publishing: 726]  [Article Influence: 36.3]  [Reference Citation Analysis (0)]
57.  Buss EC, Ho AD. Leukemia stem cells. Int J Cancer. 2011;129:2328-2336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
58.  Becker MW, Jordan CT. Leukemia stem cells in 2010: current understanding and future directions. Blood Rev. 2011;25:75-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
59.  Arai F, Suda T. Regulation of hematopoietic stem cells in the osteoblastic niche. Adv Exp Med Biol. 2007;602:61-67.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Raaijmakers MH. Niche contributions to oncogenesis: emerging concepts and implications for the hematopoietic system. Haematologica. 2011;96:1041-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
61.  Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, Nagler A, Ben-Hur H, Many A, Shultz L. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1377]  [Cited by in F6Publishing: 1330]  [Article Influence: 53.2]  [Reference Citation Analysis (0)]
62.  Avigdor A, Goichberg P, Shivtiel S, Dar A, Peled A, Samira S, Kollet O, Hershkoviz R, Alon R, Hardan I. CD44 and hyaluronic acid cooperate with SDF-1 in the trafficking of human CD34+ stem/progenitor cells to bone marrow. Blood. 2004;103:2981-2989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 413]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
63.  Lapidot T, Dar A, Kollet O. How do stem cells find their way home. Blood. 2005;106:1901-1910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 722]  [Cited by in F6Publishing: 700]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
64.  Rombouts EJ, Pavic B, Löwenberg B, Ploemacher RE. Relation between CXCR-4 expression, Flt3 mutations, and unfavorable prognosis of adult acute myeloid leukemia. Blood. 2004;104:550-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 224]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
65.  Spoo AC, Lübbert M, Wierda WG, Burger JA. CXCR4 is a prognostic marker in acute myelogenous leukemia. Blood. 2007;109:786-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 251]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
66.  Nervi B, Ramirez P, Rettig MP, Uy GL, Holt MS, Ritchey JK, Prior JL, Piwnica-Worms D, Bridger G, Ley TJ. Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100. Blood. 2009;113:6206-6214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 396]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
67.  Lane SW, Wang YJ, Lo Celso C, Ragu C, Bullinger L, Sykes SM, Ferraro F, Shterental S, Lin CP, Gilliland DG. Differential niche and Wnt requirements during acute myeloid leukemia progression. Blood. 2011;118:2849-2856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 120]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
68.  Reuss-Borst MA, Klein G, Waller HD, Müller CA. Differential expression of adhesion molecules in acute leukemia. Leukemia. 1995;9:869-874.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med. 2006;12:1167-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 891]  [Cited by in F6Publishing: 866]  [Article Influence: 48.1]  [Reference Citation Analysis (0)]
70.  Matsunaga T, Takemoto N, Sato T, Takimoto R, Tanaka I, Fujimi A, Akiyama T, Kuroda H, Kawano Y, Kobune M. Interaction between leukemic-cell VLA-4 and stromal fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia. Nat Med. 2003;9:1158-1165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 416]  [Cited by in F6Publishing: 414]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
71.  Colmone A, Amorim M, Pontier AL, Wang S, Jablonski E, Sipkins DA. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science. 2008;322:1861-1865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 436]  [Cited by in F6Publishing: 449]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]