Noncellular Niche Influences: Nervous System Mediators, Metabolic Mediators, and Hypoxia/Oxidative Stress

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. SCI-26-SCI-26
Author(s):  
Simón Méndez-Ferrer

Hematopoietic stem cells (HSCs) traffic between bone marrow and circulation, what allows for life-saving clinical transplantation. Our previous work has shown that HSC numbers in blood follow circadian oscillations that are regulated by the central pacemaker in the brain, which reaches bone marrow nestin+ mesenchymal stem cells through peripheral sympathetic nerves. In the perinatal bone marrow, HSC-niche forming mesenchymal stem cells might be different from those that form the skeleton and some of them might be neural crest-derived, like peripheral neurons and supporting glial cells. Thus, tight regulation of the bone marrow stem-cell niche in vertebrates might build upon developmental relationships of its cellular components. We have found recently that cholinergic nerves regulate HSC maintenance, proliferation and migration in divergent niches. We will present unpublished evidence of how both branches of the autonomic nervous system cooperate to regulate HSC maintenance and function in spatially and temporally distinct niches. Moreover, we have shown recently that damage to this regulatory network is essential for the manifestation of myeloproliferative neoplasms. In these diseases, previously thought to be driven solely by mutated HSCs, protecting the HSC niche might represent a novel therapeutic strategy. Patients with myeloproliferative neoplasms have a higher risk of developing acute leukemia. However, at this stage, leukemic cells might be less sensitive to the normal control by the microenvironment and, instead, acute myelogenous leukemic cells might transform the bone marrow niches to support their own survival. We will discuss potential contributions of HSC niches to myeloproliferative neoplasms and MLL-AF9-driven acute myeloid leukemia. Disclosures Off Label Use: Potential use of selective estrogen receptor modulators and beta3-adrenergic agonists in myeloproliferative neoplasms.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4-4 ◽  
Author(s):  
Simon Mendez-Ferrer ◽  
Grigori N. Enikolopov ◽  
Sergio Lira ◽  
Paul S. Frenette

Abstract The identity of mesenchymal stem cells (MSCs) and their relationship to hematopoietic stem cells (HSCs) remain poorly defined. In addition, there are discrepancies regarding the cellular constituents of the HSC niche, with studies suggesting a role for bone-lining osteoblasts, and other data implicating sinusoidal endothelial and adventitial reticular cells. Previous work from our group has demonstrated that the sympathetic nervous system (SNS) is critical for both physiological and enforced egress of HSCs from the bone marrow (BM). HSC mobilization induced by G-CSF requires signals from the SNS (Katayama et al. 2006; Cell124:407–21). Physiological release of HSCs into the bloodstream follows circadian oscillations governed by the molecular clock and triggered by cyclical norepinephrine secretion by the SNS in the BM, activation of the β3-adrenergic receptor (encoded in Adrb3), degradation of Sp1 transcription factor and downregulation of Cxcl12 (Mendez-Ferrer et al. 2008; Nature452:442–7). Here, we have identified the cell targeted by the SNS in the BM as a perivascular stromal cell expressing Nestin, an intermediate filament protein characteristic of neuroectoderm-derived stem cells. Using transgenic mice expressing GFP under the regulatory elements of the Nestin promoter, we show that virtually all catecholaminergic fibers in the BM are associated with Nestin+ cells, which represent 4.0 ± 0.6% of BM CD45− cells and 0.08 ± 0.01% of total BM nucleated cells, as determined by FACS analyses. Quantitative real-time PCR (QPCR) analyses have revealed a ~30-fold higher expression of the gene encoding the chemokine CXCL12 in Nestin+ cells than in the rest of BM CD45− cells, whereas Adrb3 was exclusively expressed in Nestin+ cells and not detectable in Nestin− CD45− cells. Detailed immunofluorescence analyses of the spatial distribution of HSCs in longitudinal BM sections revealed that 60% of CD150+ CD48−/Lineage− cells were directly attached to Nestin+ cells, and 90% of HSCs were located within 5 cell diameters from Nestin+ cells in the endosteal or sinusoidal regions of the BM (n=30). In long-term BM cultures, Nestin+ cells were rare, but located near HSCs/progenitors-enriched cobblestone-forming areas. BM Nestin+ cells were associated with HSCs not only physically but also functionally, because core HSC retention signals (Cxcl12, Kitl, Vcam1, Angpt1, Il7) were highly expressed by Nestin+ cells and significantly downregulated during G-CSF-induced mobilization, whereas the expression of the same genes was significantly lower and was not downregulated by G-CSF in Nestin− CD45− cells, as measured by QPCR. A non-selective β- or a selective β3-adrenergic receptor agonist also downregulated these core HSC retention genes, underscoring the role of the SNS in regulating HSC adhesion in the BM niche. Cell sorting of Nestin+ CD45− and Nestin− CD45− cells revealed that all the mesenchymal progenitor activity of the bone marrow (CFU-F) was contained in the Nestin+ cell fraction. Further, Nestin+ cells could robustly differentiate into osteoblasts and adipocytes. Lineage-tracing studies using a Nestin-CRE transgenic line bred to R26R reporter mice have confirmed the contribution of Nestin+ cells to osteoblasts and chondrocytes during development. G-CSF, which induces proliferation of hematopoietic cells in the BM at the expense of non-hematopoietic lineages, significantly downregulated markers of osteoblastic and adipogenic differentiation in BM Nestin+ CD45− cells but not in Nestin− CD45− cells. By contrast, daily administration of parathyroid hormone over five weeks, a treatment previously shown to expand both the osteoblastic and HSC pools, induced proliferation of Nestin+ cells and favored their differentiation into Col1a1-LacZ+ osteoblasts. Finally, we have found that Nestin+ CD45− cells, but not Nestin− CD45− cells, can form self-renewing spheres in clonal density culture, with a frequency similar to other neural crest-derived stem cells. After two weeks in culture, clonal spheres showed spontaneous multilineage differentiation into adipocytes and Col1a1-LacZ+ osteoblasts. Altogether, these results suggest that the HSC niche is composed of a heterotypic MSC-HSC pairing that is tightly regulated by the SNS. This association may reconcile divergent views regarding the vascular and osteoblastic locations of the HSC niche, and its regulation by the SNS might explain the crosstalk between hematopoietic and mesenchymal lineages in the BM during health and disease.


2021 ◽  
Vol 22 (15) ◽  
pp. 8166
Author(s):  
Natalia-Del Pilar Vanegas ◽  
Paola Fernanda Ruiz-Aparicio ◽  
Gloria Inés Uribe ◽  
Adriana Linares-Ballesteros ◽  
Jean-Paul Vernot

Leukemic cell growth in the bone marrow (BM) induces a very stressful condition. Mesenchymal stem cells (MSC), a key component of this BM niche, are affected in several ways with unfavorable consequences on hematopoietic stem cells favoring leukemic cells. These alterations in MSC during B-cell acute lymphoblastic leukemia (B-ALL) have not been fully studied. In this work, we have compared the modifications that occur in an in vitro leukemic niche (LN) with those observed in MSC isolated from B-ALL patients. MSC in this LN niche showed features of a senescence process, i.e., altered morphology, increased senescence-associated β-Galactosidase (SA-βGAL) activity, and upregulation of p53 and p21 (without p16 expression), cell-cycle arrest, reduced clonogenicity, and some moderated changes in stemness properties. Importantly, almost all of these features were found in MSC isolated from B-ALL patients. These alterations rendered B-ALL cells susceptible to the chemotherapeutic agent dexamethasone. The senescent process seems to be transient since when leukemic cells are removed, normal MSC morphology is re-established, SA-βGAL expression is diminished, and MSC are capable of re-entering cell cycle. In addition, few cells showed low γH2AX phosphorylation that was reduced to basal levels upon cultivation. The reversibility of the senescent process in MSC must impinge important biological and clinical significance depending on cell interactions in the bone marrow at different stages of disease progression in B-ALL.


eLife ◽  
2014 ◽  
Vol 3 ◽  
Author(s):  
Joan Isern ◽  
Andrés García-García ◽  
Ana M Martín ◽  
Lorena Arranz ◽  
Daniel Martín-Pérez ◽  
...  

Mesenchymal stem cells (MSCs) and osteolineage cells contribute to the hematopoietic stem cell (HSC) niche in the bone marrow of long bones. However, their developmental relationships remain unclear. In this study, we demonstrate that different MSC populations in the developing marrow of long bones have distinct functions. Proliferative mesoderm-derived nestin− MSCs participate in fetal skeletogenesis and lose MSC activity soon after birth. In contrast, quiescent neural crest-derived nestin+ cells preserve MSC activity, but do not generate fetal chondrocytes. Instead, they differentiate into HSC niche-forming MSCs, helping to establish the HSC niche by secreting Cxcl12. Perineural migration of these cells to the bone marrow requires the ErbB3 receptor. The neonatal Nestin-GFP+ Pdgfrα− cell population also contains Schwann cell precursors, but does not comprise mature Schwann cells. Thus, in the developing bone marrow HSC niche-forming MSCs share a common origin with sympathetic peripheral neurons and glial cells, and ontogenically distinct MSCs have non-overlapping functions in endochondrogenesis and HSC niche formation.


Blood ◽  
1984 ◽  
Vol 64 (6) ◽  
pp. 1288-1291 ◽  
Author(s):  
L Glasser ◽  
LB Somberg ◽  
WR Vogler

Abstract Autologous bone marrow transplantation is potentially curative in the treatment of acute leukemia if residual leukemic cells in the marrow can be eliminated prior to transplantation. We studied the purging effects of a synthetic alkyl-lysophospholipid (ALP) on marrow containing leukemic cells from a transplantable myelomonocytic leukemia (WEHI-3B) in BALB/c mice. Simulated remission bone marrow containing 2% leukemic cells treated in vitro with 20 and 100 micrograms/mL of ET-18- OCH3 (1-octadecyl-2-methyl-sn-glycerol-3-phosphocholine) significantly prolonged survival of lethally irradiated transplanted recipients. At a dose of 100 micrograms/mL, 88% of the mice survived for the duration of the experiment (approximately five months). Autopsies showed that 25% of these survivors had microscopic evidence of leukemia. Thus, in vitro treatment of marrow eliminated leukemic blasts and spared sufficient normal stem cells to allow hematologic reconstitution. The effect of ET- 18-OCH3 is not entirely selective for leukemic cells. A spleen colony assay showed that ALP has some cytotoxic effect on normal hematopoietic stem cells.


Cells ◽  
2020 ◽  
Vol 9 (1) ◽  
pp. 221
Author(s):  
Yu-Hee Kim ◽  
Kyung-Ah Cho ◽  
Hyun-Ji Lee ◽  
Minhwa Park ◽  
Sang-Jin Shin ◽  
...  

Cotransplantation of mesenchymal stem cells (MSCs) with hematopoietic stem cells (HSCs) has been widely reported to promote HSC engraftment and enhance marrow stromal regeneration. The present study aimed to define whether MSC conditioned medium could recapitulate the effects of MSC cotransplantation. Mouse bone marrow (BM) was partially ablated by the administration of a busulfan and cyclophosphamide (Bu–Cy)-conditioning regimen in BALB/c recipient mice. BM cells (BMCs) isolated from C57BL/6 mice were transplanted via tail vein with or without tonsil-derived MSC conditioned medium (T-MSC CM). Histological analysis of femurs showed increased BM cellularity when T-MSC CM or recombinant human pleiotrophin (rhPTN), a cytokine readily secreted from T-MSCs with a function in hematopoiesis, was injected with BMCs. Microstructural impairment in mesenteric and BM arteriole endothelial cells (ECs) were observed after treatment with Bu–Cy-conditioning regimen; however, T-MSC CM or rhPTN treatment restored the defects. These effects by T-MSC CM were disrupted in the presence of an anti-PTN antibody, indicating that PTN is a key mediator of EC restoration and enhanced BM engraftment. In conclusion, T-MSC CM administration enhances BM engraftment, in part by restoring vasculature via PTN production. These findings highlight the potential therapeutic relevance of T-MSC CM for increasing HSC transplantation efficacy.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2335-2335
Author(s):  
Iekuni Oh ◽  
Akira Miyazato ◽  
Hiroyuki Mano ◽  
Tadashi Nagai ◽  
Kazuo Muroi ◽  
...  

Abstract Mesenchymal stem cells (MSCs) account for a very small population in bone marrow stroma as a non-hematopoietic component with multipotency of differentiation into adipocytes, osteocytes and chondrocytes. MSC-derived cells are known to have hematopoiesis-supporting and immunomodulatory abilities. Although clinical applications of MSCs have already been conducted for the suppression of graft versus host disease in allogeneic stem cell transplantation and for tissue regeneration, underlying mechanisms of the biological events are still obscure. Previously, we established a differentiation model of MSCs using a mouse embryo fibroblast cell line, C3H10T1/2 (10T1/2) (Nishikawa M et al: Blood81:1184–1192, 1993). Preadipocyte (A54) and myoblast (M1601) cell lines were cloned by treatment with 5-azacytidine. A54 cells and M1601 cells can terminally differentiate into adipocytes and myotubes, respectively, under appropriate conditions, while parent 10T1/2 cells remain undifferentiated. Moreover, A54 cells show a higher ability to support hematopoiesis compared with the other cell lines. In this study, we analyzed gene expression profiles of the three cell lines by using DNA microarray and real-time PCR to investigate molecular mechanisms for maintaining immaturity of parent 10T1/2 cells. In A54 cells, 202 genes were up-regulated, including those encoding critical factors for hematopoiesis such as SCF, Angiopoietin-1, and SDF-1 as well as genes known to be involved in adipocyte differentiation such as C/EBPα, C/EBPδ and PPAR-γ genes. These data are consistent with the hematopoiesis-supporting ability of A54 cells. During adipocyte differentiation, SCF and SDF-1 expression levels decreased in A54 cells while C/EBPα expression showed a steady level. Recently, osteoblasts have been reported to play crucial roles in “niche” for self-renewal of hematopoietic stem cells. Our results also implicate that precursor cells of non-hematopoietic components may have important roles for hematopoiesis in bone marrow. Meanwhile, in parent 10T1/2 cells, 105 genes were up-regulated, including CD90, Dlk, Wnt5α and many functionally unknown genes. Although C/EBPα expression was induced in 10T1/2 cells without differentiation under the adipocyte differentiation conditions, CD90 expression decreased, Dlk showed a steady level and Wnt5α was up-regulated. Assuming that some regulatory mechanisms are needed to keep an immature state of parent 10T1/2 cells even under the differentiation-inducible conditions, we performed following experiments. First, enforced Dlk expression in A54 cells did not inhibit terminal differentiation to adipocytes under the differentiation conditions. Second, when we cultured A54 cells in the conditioned media of parent 10T1/2 cells under the differentiation-inducible conditions, adipocyte differentiation was inhibited, suggesting that 10T1/2 cells produce some soluble molecules that can inhibit adipocyte differentiation. Since Wnt family is known to be involved in the regulation of self-renewal of several stem cells, Wnt5α may be one candidate for maintenance of “stemness” of MSCs. Taken together, the data of 10T1/2 cells suggest that MSCs can self-regulate their differentiation in the bone marrow stromal system. This concept may be important to investigate the fatty change of bone marrow in aging and in aplastic anemia.


Sign in / Sign up

Export Citation Format

Share Document