Functional and Molecular Alterations of Bone Marrow Mesenchymal Stem and Progenitor Cells in Patients with Myelodysplastic Syndrome with Ring Sideroblast

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1489-1489
Author(s):  
Monika Dolinska ◽  
Pingnan Xiao ◽  
Anne-Sofie Johansson ◽  
Lakshmi Sandhow ◽  
Makoto Kondo ◽  
...  

Abstract Myelodysplastic Syndrome with Ring Sideroblast (MDS-RS), a clonal hematopoietic cell neoplasm, is low risk MDS, characterized by anemia, hyperplastic ineffective erythropoiesis and marrow ring sideroblasts. Mouse studies have shown that bone marrow niche, including endothelial cells, osteoblasts, adipocytes and mesenchymal stem cells (MSCs), contribute to progression of various hematological disorders. However, in vivo contribution of the different bone marrow stromal cells to the progression of MDS-RS in patients remains largely unknown. To investigate this, we have phenotypically, molecularly and functionally characterized the BM native stromal cell subsets including MSCs freshly isolated by multi-color fluorescence activated cell sorting (FACS) from bone marrow of MDS-RS patients and age-matched healthy donors. We found: 1) the MDS-RS MSCs, estimated by colony forming unit-fibroblast (CFU-F), shared similar immunophenotype with normal MSCs (CD45-CD235a-CD31-CD44-, most of which were CD271+CD146+CD106+); 2) the frequency of CFU-Fs was significantly increased in the phenotypically defined MSCs of MDS-RS bone marrow compared to that of age-matched healthy controls (p=0.005); 3) multi-lineage differentiation assay revealed impaired osteogenic differentiation potential, but enhanced adipogenic differentiation potential of MDS-RS MSCs; 4) FACS analysis showed increased frequency of the adhesion receptor integrin α4 (ITGA4) in the CD44- MSCs from MDS-RS bone marrow (p=0.013); 5) Correspondingly, RNA-sequencing of the freshly isolated bone marrow MSCs and endothelial cells revealed altered gene expression profile of these cells in MDS-RS patients. Among those, ITGA4, ITGA11, ITGAE and ITGB1 are upregulated in the MDS-RS MSCs, indicating potential abnormal adhesive interaction of the MSCs with hematopoietic stem cells in the patients. In addition, the cell cycling gene KI67 is upregulated whereas cell cycle negative regulators, like CDKN1A and CDKN1C are downregulated in the MDS-RS MSCs, which is consistent with their increased CFU-F activity. Interestingly, we detected abnormal expression of hematopoietic growth factors such as downregulation of ANGPTL4 in the MDS-RS MSCs and upregulation of ANGPT1 in the MDS-RS endothelial cells. The functional relationship between the stromal cell alterations and the abnormal hematopoiesis as well as the underlying molecular mechanisms are currently under investigation. Taken together, our data provide new evidence for phenotypic, functional and molecular alterations of bone marrow mesenchymal cells in MDS-RS patients. The molecular pathways mediating bone marrow niche alteration could be potential therapeutic targets for new treatment of MDS-RS. Disclosures No relevant conflicts of interest to declare.

Author(s):  
Xiaofang Chen ◽  
Ningyu Li ◽  
Jianyu Weng ◽  
Xin Du

Myelodysplastic syndrome (MDS) is a group of clonal hematopoietic disorders related to hematopoietic stem and progenitor cell dysfunction. However, therapies that are currently used to target hematopoietic stem cells are not effective. These therapies are able to slow the evolution toward acute myeloid leukemia but cannot eradicate the disease. Mesenchymal stem cells (MSCs) have been identified as one of the main cellular components of the bone marrow microenvironment, which plays an indispensable role in normal hematopoiesis. When functional and regenerative capacities of aging MSCs are diminished, some enter replicative senescence, which promotes inflammation and disease progression. Recent studies that investigated the contribution of bone marrow microenvironment and MSCs to the initiation and progression of the disease have offered new insights into the MDS. This review presents the latest updates on the role of MSCs in the MDS and discusses potential targets for the treatment of MDS.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2398-2398
Author(s):  
Monika Dolinska ◽  
Johannis Klang ◽  
Pingnan Xiao ◽  
Andranik Durgaryan ◽  
Lakshmi Sandhow ◽  
...  

Abstract Chronic myeloid leukemia (CML) is a myeloproliferative stem cell neoplasm characterized by the presence of the BCR-ABL1 fusion gene. Although current treatment with tyrosine kinase inhibitors (TKI) has dramatically improved the prognosis of CML, these inhibitors do not eradicate leukemic stem cells (LSC) in most patients with the risk of recurrence of leukemia if TKI are stopped. In vitro studies have suggested that this might be attributable to protection of bone marrow (BM) stromal cells, such as osteoblasts, adipocytes, endothelial and mesenchymal stem cells (MSCs). However, how different BM stromal cells contribute to the persistence of LSC remains largely unknown. To investigate this issue we have compared freshly isolated BM stromal cell subsets including MSCs from newly diagnosed CML patients (n=10) with that from age-matched healthy donors (n=12). Distinct from the previous studies on culture-selected BM stromal cells, the naive stromal cells isolated by multi-color fluorescence activated cell sorting (FACS) were phenotypically, molecularly and functionally characterized in the present study. We observed: 1) Similar to the immunophenotype of normal MSCs (CD45-CD235a-CD31-CD44-, most of which were CD271+CD146+CD106+) (Qian et al., JBC, 2012), the CML MSCs, estimated by colony forming unit-fibroblast (CFU-F), were also enriched in the CD45-CD235a-CD31-CD44- cell fraction. 2) The frequency of CFU-Fs was significantly increased in CML BM compared to that in the age-matched healthy controls (p=0.005). 3) A decreased osteogenic, but enhanced adipogenic differentiation potential of CML MSC was revealed in multilineage differentiation assay. This suggests a skewed differentiation potential of the CML MSCs towards adipocytes, possibly related to an altered stromal cell composition in the patients; 4) An increased proportion of CD31+ endothelial cells was seen in CML BM stroma compared to controls (p=0.023) by FACS. 5) An upregulation of the adhesion receptor integrin α4/CD49D was seen in the CD44- MSCs from CML patients (p=0.0087). Conversely, a downregulation of transcripts of Angiopoietin 1, CXCL12, KIT ligand and LAMA4 in the patient MSCs was detected by Quantitative-PCR, indicating an altered hematopoiesis-supportive function of CML MSCs. 6) Importantly, no BCR-ABL fusion were found in the freshly sorted MSCs and mature stromal cells using Fluorescence In Situ Hybridization analysis, suggesting that these MSCs were not part of the leukemic clone. Taken together, our data provide evidence for phenotypic and functional alterations of BM mesenchymal cells in CML patients. The functional relationship between the stromal cell alterations and the growth of LSC as well as the underlying molecular mechanisms are currently under investigation. Disclosures Mustjoki: Finnish Cancer Institute: Research Funding; Sigrid Juselius Foundation: Research Funding; Academy of Finland: Research Funding; the Finnish Cancer Societies: Research Funding; Pfizer: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Signe and Ane Gyllenberg Foundation: Research Funding.


Author(s):  
Basem M. Abdallah ◽  
Hany M. Khattab

: The isolation and culture of murine bone marrow-derived mesenchymal stromal stem cells (mBMSCs) have attracted great interest in terms of the pre-clinical applications of stem cells in tissue engineering and regenerative medicine. In addition, culturing mBMSCs is important for studying the molecular mechanisms of bone remodelling using relevant transgenic mice. Several factors have created challenges in the isolation and high-yield expansion of homogenous mBMSCs; these factors include low frequencies of bone marrow-derived mesenchymal stromal stem cells (BMSCs) in bone marrow, variation among inbred mouse strains, contamination with haematopoietic progenitor cells (HPCs), the replicative senescence phenotype and cellular heterogeneity. In this review, we provide an overview of nearly all protocols used for isolating and culturing mBMSCs with the aim of clarifying the most important guidelines for culturing highly purified mBMSC populations retaining in vitro and in vivo differentiation potential.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Marie-Theresa Weickert ◽  
Judith S. Hecker ◽  
Michèle C. Buck ◽  
Christina Schreck ◽  
Jennifer Rivière ◽  
...  

AbstractMyelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal hematopoietic stem cell disorders with a poor prognosis, especially for elderly patients. Increasing evidence suggests that alterations in the non-hematopoietic microenvironment (bone marrow niche) can contribute to or initiate malignant transformation and promote disease progression. One of the key components of the bone marrow (BM) niche are BM stromal cells (BMSC) that give rise to osteoblasts and adipocytes. It has been shown that the balance between these two cell types plays an important role in the regulation of hematopoiesis. However, data on the number of BMSC and the regulation of their differentiation balance in the context of hematopoietic malignancies is scarce. We established a stringent flow cytometric protocol for the prospective isolation of a CD73+ CD105+ CD271+ BMSC subpopulation from uncultivated cryopreserved BM of MDS and AML patients as well as age-matched healthy donors. BMSC from MDS and AML patients showed a strongly reduced frequency of CFU-F (colony forming unit-fibroblast). Moreover, we found an altered phenotype and reduced replating efficiency upon passaging of BMSC from MDS and AML samples. Expression analysis of genes involved in adipo- and osteogenic differentiation as well as Wnt- and Notch-signalling pathways showed significantly reduced levels of DLK1, an early adipogenic cell fate inhibitor in MDS and AML BMSC. Matching this observation, functional analysis showed significantly increased in vitro adipogenic differentiation potential in BMSC from MDS and AML patients. Overall, our data show BMSC with a reduced CFU-F capacity, and an altered molecular and functional profile from MDS and AML patients in culture, indicating an increased adipogenic lineage potential that is likely to provide a disease-promoting microenvironment.


Blood ◽  
2007 ◽  
Vol 110 (1) ◽  
pp. 82-90 ◽  
Author(s):  
Younghun Jung ◽  
Jingcheng Wang ◽  
Junhui Song ◽  
Yusuke Shiozawa ◽  
Jianhua Wang ◽  
...  

Differentiation of hematopoietic stem cells (HSCs) after birth is largely restricted to the bone marrow cavity, where HSCs are associated closely with osteoblasts (OBs). How OBs localize HSCs to the endosteal niche remains unclear. To explore adhesive interactions between HSCs and OBs, a cell blot analysis was used that revealed 2 major bands that corresponded to monomers and multimers of annexin II (Anxa2). Immunohistochemistry revealed that OBs and marrow endothelial cells express Anxa2 at high levels. Function-blocking studies confirmed that Anxa2 mediates HSC adhesion mainly via the N-terminal portion of the Anxa2 peptide. Adhesion of HSCs to OBs derived from Anxa2-deficient animals (Anxa2−/−) was significantly impaired compared with OBs obtained from wild-type animals (Anxa2+/+). Moreover, fewer HSCs were found in the marrow of Anxa2−/− versus Anxa2+/+ animals. Short-term lodging, engraftment, and survival of irradiated mice with whole marrow cells were substantially inhibited by N-terminal peptide fragments of Anxa2 or anti-Anxa2 antibodies. Similar findings were noted in long-term competitive repopulation studies. Collectively, these findings reveal that Anxa2 regulates HSC homing and binding to the bone marrow microenvironment and suggest that Anxa2 is crucial for determining the bone marrow niche of HSCs.


2019 ◽  
Vol 141 (7-8) ◽  
pp. 233-237

Myelodysplastic syndrome (MDS) is a clonal hematopoietic stem cell disorder characterized by ineffective hematopoiesis and cytopenia in peripheral blood, where about a third of patients may develop acute myeloid leukemia (AML). The diagnosis of MDS requires the analysis of peripheral blood and bone marrow. Depending on the percentage of blasts in the bone marrow, the number of cytopenias and cytogenetic abnormalities, determination of the prognostic indices is possible (IPSS – „International Prognostic Scoring System“, R-IPSS-„Revised International Prognostic Scoring System“, WPSS – „WHO Prognostic Scoring System“). Until today, numerous studies have been conducted on the molecular mechanisms and epigenetic pathways in myelodysplastic syndrome, and their prognostic and therapeutic importance, but there are few studies analyzing the importance of microRNAs (miRNAs) in MDS. In the last few years, there have been numerous results on the impact of aberrant miRNA expression in malignant disorders where the miRNA represent tumor suppressor genes or oncogenes. Several miRNAs have been recognized as diagnostic and prognostic parameters and possible therapeutic targets. In this paper, we present the overview of recent results on the role of miRNA in MDS.


2016 ◽  
Vol 364 (3) ◽  
pp. 573-584 ◽  
Author(s):  
Patrick Wuchter ◽  
Rainer Saffrich ◽  
Stefan Giselbrecht ◽  
Cordula Nies ◽  
Hanna Lorig ◽  
...  

2020 ◽  
Vol 2020 ◽  
pp. 1-12
Author(s):  
Huihong Zeng ◽  
Jiaoqi Cheng ◽  
Ying Fan ◽  
Yingying Luan ◽  
Juan Yang ◽  
...  

Development of hematopoietic stem cells is a complex process, which has been extensively investigated. Hematopoietic stem cells (HSCs) in mouse fetal liver are highly expanded to prepare for mobilization of HSCs into the fetal bone marrow. It is not completely known how the fetal liver niche regulates HSC expansion without loss of self-renewal ability. We reviewed current progress about the effects of fetal liver niche, chemokine, cytokine, and signaling pathways on HSC self-renewal, proliferation, and expansion. We discussed the molecular regulations of fetal HSC expansion in mouse and zebrafish. It is also unknown how HSCs from the fetal liver mobilize, circulate, and reside into the fetal bone marrow niche. We reviewed how extrinsic and intrinsic factors regulate mobilization of fetal liver HSCs into the fetal bone marrow, which provides tools to improve HSC engraftment efficiency during HSC transplantation. Understanding the regulation of fetal liver HSC mobilization into the fetal bone marrow will help us to design proper clinical therapeutic protocol for disease treatment like leukemia during pregnancy. We prospect that fetal cells, including hepatocytes and endothelial and hematopoietic cells, might regulate fetal liver HSC expansion. Components from vascular endothelial cells and bones might also modulate the lodging of fetal liver HSCs into the bone marrow. The current review holds great potential to deeply understand the molecular regulations of HSCs in the fetal liver and bone marrow in mammals, which will be helpful to efficiently expand HSCs in vitro.


2013 ◽  
Vol 2013 ◽  
pp. 1-11 ◽  
Author(s):  
Claudia Cicione ◽  
Emma Muiños-López ◽  
Tamara Hermida-Gómez ◽  
Isaac Fuentes-Boquete ◽  
Silvia Díaz-Prado ◽  
...  

Background. The interests in mesenchymal stem cells (MSCs) and their application in cell therapy have resulted in a better understanding of the basic biology of these cells. Recently hypoxia has been indicated as crucial for complete chondrogenesis. We aimed at analyzing bone marrow MSCs (BM-MSCs) differentiation capacity under normoxic and severe hypoxic culture conditions.Methods. MSCs were characterized by flow cytometry and differentiated towards adipocytes, osteoblasts, and chondrocytes under normoxic or severe hypoxic conditions. The differentiations were confirmed comparing each treated point with a control point made of cells grown in DMEM and fetal bovine serum (FBS).Results. BM-MSCs from the donors displayed only few phenotypical differences in surface antigens expressions. Analyzing marker genes expression levels of the treated cells compared to their control point for each lineage showed a good differentiation in normoxic conditions and the absence of this differentiation capacity in severe hypoxic cultures.Conclusions. In our experimental conditions, severe hypoxia affects thein vitrodifferentiation potential of BM-MSCs. Adipogenic, osteogenic, and chondrogenic differentiations are absent in severe hypoxic conditions. Our work underlines that severe hypoxia slows cell differentiation by means of molecular mechanisms since a decrease in the expression of adipocyte-, osteoblast-, and chondrocyte-specific genes was observed.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2460-2460 ◽  
Author(s):  
Hairui Su ◽  
Szu-Mam Liu ◽  
Chiao-Wang Sun ◽  
Mark T. Bedford ◽  
Xinyang Zhao

Protein arginine methylation is a common type of post-translational modification. PRMT1, the major type I protein arginine methyltransferase, catalyzes the formation of asymmetric dimethyl-arginine and is implicated in various cellular processes, including hematopoiesis and tumorigenesis. We have shown that PRMT1 expression is naturally low in hematopoietic stem cells (HSCs). However, the functions of PRMT1 in hematopoietic stem cell self-renewal and differentiation are yet to be revealed. We have found a cyanine-based fluorescent probe (E84) that can specifically label PRMT1 protein. E84 staining dynamically captures intracellular PRMT1 level and was used to separate live HSC populations with differential PRMT1 expression by flow cytometry. Subsequent bone marrow transplantation of E84high or E84low Lin−Sca1+cKit+ (LSK) cells showed that E84low LSK cells were much more advantageous in reconstituting each blood cell lineages, compared to the E84high counterparts, meaning that the stem-ness of HSCs is negatively correlated with endogenous PRMT1. Therefore, inhibition of PRMT1 was expected to enhance the number and differentiation potential of functional HSCs. The treatment of a PRMT1-specific inhibitor (MS023) to mice resulted in an enlarged LT-HSC population in bone marrow and decreased frequency of granulocyte progenitor cells. In vitro colony formation assays further demonstrated that PRMT1 is required for GMP differentiation. Then we asked whether copious expression of PRMT1 promotes the differentiation of HSC. In this line, we made a LoxP-STOP-LoxP-PRMT1 transgenic mouse model, which induces PRMT1 overexpression upon the expression of Cre recombinase from tissue-specific promoters. We established Mx1-Cre-PRMT1 (Mx1-Tg) mice. Mx1-Tg mice were injected with poly(I:C) for PRMT1 induction and analyzed at four weeks after the last dose. We found that, as predicted, LT-HSC population was reduced and the Pre-GM population was raised. Accordingly, more CFU-Gs but less GEMMs were grown on CFU assays. We further utilized this animal model to compare the blood reconstitution capabilities of bone marrow cells from Mx1-Tg vs. WT mice in the same repopulating conditions. We performed competitive bone marrow transplantation by injecting Mx1-Tg/WT (CD45.2) bone marrow plus supporting cells (CD45.1) to irradiated mice, followed by 5 doses of poly(I:C) induction. Recipient mice were analyzed during a course of approximately 16 weeks. Mx1-Tg cells were outcompeted by WT cells in reconstituting every blood lineages. Taken together, we conclude that PRMT1 promotes HSC differentiation and accelerates HSC exhaustion during the stress caused by bone marrow irradiation. To understand the mechanism on PRMT1-mediated stress hematopoiesis, we also made Pf4-Cre PRMT1 transgenic mice. When PRMT1 is specifically expressed in MK cells, the number of LT-HSCs was also reduced, implying that PRMT1 affects the self-renewal of LT-HSCs via communication between MK cells and HSCs. Mechanistically, two PRMT1 substrates - RBM15 and DUSP4 - are critical for stem cell self-renewal. We further characterized how PRMT1 activates p38 kinase pathway via directly methylating DUSP4 thus induces ubiquitylation and degradation of DUSP4. The arginine methylation site on DUSP4 has been identified. Moreover, introducing methyl-R mutated DUSP4 back to PRMT1-overexpressing cells partially rescued the loss of HSC differentiation potential. This data adds a new link between arginine methylation and protein phosphorylation mediated by MAP kinases/phosphatases. In addition, we discovered that RBM15 controls alternative RNA splicing and RNA processing in a PRMT1-dosage dependent manner. In this report, we will further address how RBM15 target genes, such as enzymes involved in fatty acid metabolic pathways, affect HSC differentiation. In summary, we report that arginine methylation is a novel regulator for the HSC differentiation via controlling p38-regulated stress pathway and metabolic reprogramming. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document