Elsevier

Leukemia Research

Volume 39, Issue 10, October 2015, Pages 1020-1027
Leukemia Research

Invited review
Bone marrow niche in the myelodysplastic syndromes

https://doi.org/10.1016/j.leukres.2015.06.017Get rights and content

Highlights

  • We discuss important stomal cells in the MDS BM niche.

  • We discuss important cytokines in the MDS BM niche.

  • We Identify opportunities for further study of the MDS BM niche.

Abstract

The myelodysplastic syndromes (MDS) are a diverse group of clonal hematopoietic malignancies characterized by ineffective hematopoiesis, progressive bone marrow (BM) failure, cytogenetic and molecular abnormalities, and variable risk of progression to acute myeloid leukemia (AML). The BM microenvironment in MDS plays an important role in the development of this disorder. The BM stromal cells of MDS patients often harbor distinct chromosomal aberrations than the hematopoietic elements, suggesting different genetic origins. Perturbed cytokine secretions from BM stromal cells such as multipotent mesenchymal stem cells (MSCs) and endothelial cells are associated with increased proliferation and survival of malignant hematopoietic cells. Within the MDS BM there are also alterations in stromal cell composition, signaling and angiogenesis between Low- and High-risk MDS patients. Several open lines of investigation into the MDS niche remain, including the timing of stromal defects in context to dysplastic hematopoiesis. Another important, unanswered question is the impact of age on BM stroma function and regulation (or dysregulation) or hematopoietic stem/progenitor cells. With a better understanding of the MDS niche, new therapeutic strategies will emerge.

Introduction

The myelodysplastic syndromes (MDS) comprise a group of clonal hematological malignancies resulting in bone marrow (BM) failure and increased risk for progression to acute myeloid leukemia (AML) [1], [2]. Next generation DNA sequencing of hundreds of MDS patients has revealed that MDS is a highly heterogeneous multigenetic disease with sub clonal architecture [2], [3]. Recent studies of the general population found that 5–10% of older, apparently healthy individuals had acquired ≥1 myeloid gene mutation, whereas younger individuals were much less likely to have acquired clonal hematopoiesis with somatic mutations [4], [5]. Together, these results support the notion that the origin of MDS is tied to cellular senescence and provide biological rationale for why MDS most often presents in the seventh and eight decades of life [6], [7]. Clinically, MDS is often diagnosed after recognizing symptoms related to BM failure and cytopenias (e.g. fatigue, pallor, infections, bruising, bleeding). MDS has a multi-step pathogenic process. Early stages of the disease (Low to Intermediate-risk MDS) harbor rare, multipotent stem cells with somatic genomic mutations [1], [2]. This clone is associated with dysplastic hematopoiesis, excessive release of myelosuppressive cytokines, defective differentiation and genomic instability. MDS transformation to AML is a result of continuation of this process, hypermethylation, silencing of tumor suppressor genes (e.g. p15) and activation of oncogenes (e.g. Ras) [2]. Although the MDS BM is hyperproliferative, the net balance is ineffective hematopoiesis due to increased apoptosis of malignant progenitors. Apoptosis in progenitor cells is increased due to cell-intrinsic signals like BCL-2 family proteins, but BM niche signaling also promotes apoptosis via TNF-α, Fas ligand, and TGF-β [1], [3].

Whereas the majority of current MDS studies focus on genetic and epigenetic events required for normal HSC transformation into malignant hematopoietic cells, there is mounting evidence of a BM niche-based model for MDS genesis that predisposes normal HSCs to genomic mutations [4], [5].

In normal hematopoiesis, the BM niche controls hematopoietic cells via paracrine regulation, cell–cell contact and extracellular matrix (ECM) deposition. Within the BM niche, multipotent mesenchymal stromal cells (MSCs) serve an important role in regulating HSC self-renewal and differentiation [6], [7], [8]. Other important BM stromal cells include osteoblasts, osteoclasts, endothelial cells, fibroblasts, adipocytes and chondrocytes (Fig. 1) [9], [10]. Close cooperation between the BM niche and HSC balances the dynamic needs for hematopoiesis and tissue turnover [9], [11].

As for the MDS niche, the picture is ill-defined. However, emerging data provide enough concepts to allow for a framework of understanding. In a most basic frame, a cluster of atypical located immature precursors (ALIPs) physically and chemically interact with a unique orchestration of BM stromal elements (Fig. 2) [6], [7], [12].

In terms of paracrine regulation by BM stroma, the MDS niche is rife with myelotoxic cytokine imbalances compared to normal BM (Table 1) [13]. Not only do the imbalances in cytokine release depress hematopoiesis, they further perturb BM angiogenesis, ECM deposition, facilitate progressive genomic instability and contribute to the immune evasion of MDS cells [14].

Section snippets

MSCs in MDS BM niche

MSCs serve important roles in hematopoiesis and immune regulation. Several studies have indicated that impaired MSCs propagate MDS [8]. Among the MSC impairments is altered expression of Aurora kinases genes (AURK). AURKs are mitotic kinases with an important role in the regulation of G2/M phase of cell cycle, centrosomes and cytokinesis. Upregulation of AURK in cells causes dysregulation of mitosis and meiosis, which results in increased ontogenesis [14], [15]. Recent studies have indicated

The supportive role of osteoblast cells in MDS BM niche

As stromal cells in the endosteal niche, osteoblasts serve important regulatory roles in MDS BM microenvironment [24]. In addition, osteoblasts regulate the maturation and proliferation of osteoclasts through factors like receptor activator of nuclear factor kappa-B ligand (RANKL). Osteoclasts have fundamental functions in the BM niche. They are involved in HSCs support by secreting heparan sulfate, and play important roles in development and maintenance of bony skeleton of BM [28]. Osteoblasts

Vascular niche and other stromal cells in MDS bone marrow

Several types of stromal cells comprise the BM vascular niche. These are adventitial reticular CD146+ cells (ARCs) expressing Ang-1 in high levels. These cells belong to a group of osteogenic progenitors involved in development of BM heterotropic ossicles. ARCs are located near endothelial cells and are capable of direct signaling not only with HSCs but with endothelial cells, reinforcing signals to HSCs from the latter [38]. Endothelial cells play an important role in homing of hematopoietic

BM niche in low-risk and high-risk MDS

In the early stages of MDS (Low-risk MDS), increased apoptosis in hematopoietic progenitors and cytopenia is observed. In advanced stages of MDS (High risk), increased resistance to apoptosis is accompanied by further proliferation and disease transformation to AML [2], [24].

Several mechanisms of apoptosis have been studied in MDS. Apoptosis can be increased by activated T-cells because of their activity in elimination of the malignant clone, expression of Bcl-2 or reduction in hematopoietic

Next generation DNA sequencing in MDS

Next-generation sequencing (NGS) has provided new insights into the molecular etiology of MDS [56]. Early NGS studies revealed mutations of TP53, RUNX1, NRAS, and FLT3 in MDS patients, albeit at low rates. However, recent NGS studies have identified more common mutations involving epigenetic regulators such as TET2, IDH1/2, DNMT3A, ASXL1 and EZH2. In addition, multiple components of the RNA splicing machinery can be mutated in MDS, such as SF3B1, SRSF2, U2AF1, and ZRSR2. Other mutations in MDS

Conclusion and future perspective

The MDS BM microenvironment is clearly perturbed when compared to the normal BM microenvironment. The fact that MDS stromal cells often harbor different genomic mutations than their hematopoietic counterparts signals different origins. This finding also begs the question whether MDS stromal defects came first or whether the hematopoietic stem/progenitor cell defects came first. If the stromal defects came first, possibly as a consequence of cellular senescence, there are plenty of data showing

Authors’ contributions

Najmaldin Saki and Christopher R. Cogle conceived the manuscript and revised it; Elahe Khodadi and Mohammad Shahjahani wrote the manuscript; Shirin Azizidoost and June Li helped writing final version of manuscript; CRC edited the manuscript.

Conflict of interest statement

The authors declare no conflict of interest.

Acknowledgments

We wish to thank all our colleagues in Health research institute, Research Center of Thalassemia & Hemoglobinopathy. CRC was supported by a Scholar in Clinical Research Award by the Leukemia & Lymphoma Society. This work was also supported by the Gatorade Trust, which is administered by the University Of Florida Department Of Medicine.

References (62)

  • J. Zhu et al.

    Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells

    Blood

    (2007)
  • R.S. Taichman et al.

    Human osteoblasts support human hematopoietic progenitor cells in vitro bone marrow cultures

    Blood

    (1996)
  • R. Borojevic et al.

    Bone marrow stroma in childhood myelodysplastic syndrome: composition, ability to sustain hematopoiesis in vitro, and altered gene expression

    Leuk. Res..

    (2004)
  • Y. Jung et al.

    Annexin II expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation

    Blood

    (2007)
  • S.K. Nilsson et al.

    Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells

    Blood

    (2005)
  • M.J. Kiel et al.

    Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance

    Cell Stem Cell

    (2007)
  • B. Sacchetti et al.

    Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment

    Cell

    (2007)
  • E. Flores-Figueroa et al.

    In vitro characterization of hematopoietic microenvironment cells from patients with myelodysplastic syndrome

    Leuk. Res.

    (2002)
  • C. Fozza et al.

    The role of T-cells in the pathogenesis of myelodysplastic syndromes: passengers and drivers

    Leuk. Res.

    (2013)
  • D. Campioni et al.

    In vitro evaluation of bone marrow angiogenesis in myelodysplastic syndromes: a morphological and functional approach

    Leuk. Res.

    (2004)
  • W.T. Bellamy et al.

    Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes

    Blood

    (2001)
  • H. van Kamp et al.

    Clonal involvement of granulocytes and monocytes, but not of T and B lymphocytes and natural killer cells in patients with myelodysplasia: analysis by X-linked restriction fragment length polymorphisms and polymerase chain reaction of the phosphoglycerate kinase gene

    Blood

    (1992)
  • W.T. Bellamy et al.

    Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes

    Blood

    (2001)
  • A.E. Smith et al.

    Next-generation sequencing of the TET2 gene in 355 MDS and CMML patients reveals low-abundance mutant clones with early origins, but indicates no definite prognostic value

    Blood

    (2010)
  • C.R. Cogle et al.

    High rate of uncaptured myelodysplastic syndrome cases and an improved method of case ascertainment

    Leuk. Res.

    (2014)
  • D.E. Rollison et al.

    Epidemiology of myelodysplastic syndromes and chronic myeloproliferative disorders in the United States, 2001–2004, using data from the NAACCR and SEER programs

    Blood

    (2008)
  • H.G.P. Raaijmakers Marc et al.

    Bone progenitor dysfunction induces myelodysplasia and secondary leukemia

    Nature

    (2010)
  • D. Hanahan et al.

    The hallmarks of cancer

    Cell

    (2000)
  • Y. Shiozawa et al.

    The bone marrow niche: habitat to hematopoietic and mesenchymal stem cells, and unwitting host to molecular parasites

    Leukemia

    (2008)
  • Y. Miura et al.

    Mesenchymal stem cell-organized bone marrow elements: an alternative hematopoietic progenitor resource

    Stem Cells

    (2006)
  • R.S. Taichman et al.

    Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor

    J. Exp. Med.

    (1994)
  • Cited by (66)

    • Early detection and intervention of clonal hematopoiesis for preventing hematological malignancies

      2022, Cancer Letters
      Citation Excerpt :

      Bone marrow (BM) niche has also been implicated in CH possibly. For example, in MDS, the abnormality of BM stromal cells such as multipotent mesenchymal stem cells (MSCs) and endothelial cells can regulate the release of TNF- α、IL-6、IFN- γ and other cytokines [101], thus leading to CH through promoting the proliferation and survival advantage of malignant hematopoietic clones. Therefore, in similarity to the previously discussed seven factors, BM niche provides microenvironmental signals for the formation of CH.

    • Combination of dociparstat sodium (DSTAT), a CXCL12/CXCR4 inhibitor, with azacitidine for the treatment of hypomethylating agent refractory AML and MDS

      2021, Leukemia Research
      Citation Excerpt :

      Conversely, disrupting CXCR4 signaling inhibits transmigration, survival, and chemotherapy resistance in vitro and tumor growth in AML xenografts [15–17]. Likewise, in MDS, the bone marrow niche upregulates production of CXCR4 and CXCL12 and many cell surface adhesion proteins that alter homing, proliferation, and survival of malignant stem cells, protecting them from cytotoxic chemotherapy [18,19]. Heparin avidly binds to a cluster of cationic amino acids on the CXCL12 surface that overlaps with the CXCR4 binding region [20,21].

    • Myelodysplastic Syndromes

      2019, Abeloff’s Clinical Oncology
    View all citing articles on Scopus
    View full text