scholarly journals Effects of High-Mobility Group A Protein Application on Canine Adipose-Derived Mesenchymal Stem CellsIn Vitro

2012 ◽  
Vol 2012 ◽  
pp. 1-10 ◽  
Author(s):  
A. A. Ismail ◽  
S. Wagner ◽  
H. Murua Escobar ◽  
S. Willenbrock ◽  
K. A. Sterenczak ◽  
...  

Multipotency and self-renewal are considered as most important features of stem cells to persist throughout life in tissues. In this context, the role of HMGA proteins to influence proliferation of adipose-derived mesenchymal stem cell (ASCs) while maintaining their multipotent and self-renewal capacities has not yet been investigated. Therefore, extracellular HMGA1 and HMGA2 application alone (10–200 ng/mL) and in combination with each other (100, 200 ng/mL each) was investigated with regard to proliferative effects on canine ASCs (cASCs) after 48 hours of cultivation. Furthermore, mRNA expression of multipotency marker genes in unstimulated and HMGA2-stimulated cASCs (50, 100 ng/mL) was analyzed by RT-qPCR. HMGA1 significantly reduced cASCs proliferation in concentrations of 10–200 ng/mL culture medium. A combination of HMGA1 and HMGA2 protein (100 and 200 ng/mL each) caused the same effects, whereas no significant effect on cASCs proliferation was shown after HMGA2 protein application alone. RT-qPCR results showed that expression levels of marker genes including KLF4, SOX2, OCT4, HMGA2, and cMYC mRNAs were on the same level in both HMGA2-protein-stimulated and -unstimulated cASCs. Extracellular HMGA protein application might be valuable to control proliferation of cASCs in context with their employment in regenerative approaches without affecting their self-renewal and multipotency abilities.

2008 ◽  
Vol 396-398 ◽  
pp. 123-126
Author(s):  
Timothy Wilson ◽  
Reeta Viitala ◽  
Mervi Puska ◽  
Mika Jokinen ◽  
Risto Penttinen

The role of silica and macrophages in fibrosis is well documented, but in bone formation it is relatively unknown despite decades of research with bioactive glasses. In this study macrophages were isolated from rat peritoneal and then cultured for five days in the presence of two types of silica microparticles with different solubilities. After the fifth day the culture medium was collected, purified and used as an additive in bone marrow derived rat stem cell cultures. The stem cells were cultured for five days in α-mem containing only 0,5% of FCS, enabling cell survival but disrupting their proliferation. As controls, stem cells were also cultured in α-mem containing silica microparticles. At days one and five the amount of soluble collagen was assayed from the culture medium and the cells were counted. All stem cell cultures with macrophage medium additives were found to be proliferative, with statistically significant difference to controls. However, collagen was only produced in cultures containing medium from macrophages cultured with fast-dissolving silica microparticles. This suggests that silica can induce cell proliferation and extra cellular matrix protein secretion which is mediated by macrophages, and that the solubility of silica is also a major factor in this reaction.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1076-1076
Author(s):  
Severine Marti N-Lannerée ◽  
Marie-Laure Bonnet ◽  
Caroline Mayeur-Rousse ◽  
Angelina Bertrand ◽  
Marie-Claude Meunier ◽  
...  

Abstract In previous experiments we have demonstrated that BCR-ABL activates specifically STAT3 in the context of murine ES cells and in leukemic CD34+ cells in patients with chronic myelogenous leukemia. This activation occurs essentially through Tyr705 and Ser 727 phosphorylation and implicates Jak2 and MEK pathways (Coppo et al, Brit J Haematol, 2006). However, it is not known if STAT3 activation plays a role in the self-renewal of primitive stem cells or if it is predominantly involved in BCR-ABL-associated leukemogenesis. To determine the role of STAT3 at the primitive stem cell level, we have inhibited specifically STAT3 expression by using a shRNA-GFP-STAT3 construct which was lentivirally transduced into purified CD34+ cells from patients with CML. Western blot experiments determined the specificity of the shRNA-STAT3 construct in hematopoietic cell lines with specific inhibition of STAT3 with no interference with STAT1, STAT5a or STAT5b expression. 8 patients with CML at diagnosis were included in the study. CD34+ cells purified from cord blood (CB) or peripheral blood stem cell (PBCS) collects were used as controls. Each sample has been transduced with high titer lentiviruses expressing either sh-STAT3 or sh-luciferase control. After transduction, GFP+ cells were purified by cell-sorting and assayed in clonogenic assays as well as in longterm- culture assays in the presence of MS-5 stromal layers with weekly half-medium changes. At week+5, clonogenic assays were performed to evaluate the numbers of LTC-IC- derived progeny. The inhibition of STAT3 expression did not alter significantly the clonogenic cell potentials in CB-CD34+ cells (n=2) or PBSC (n=1) samples. In LTC-IC assays, STAT3 inhibition resulted in 1.8-fold reduced clonogenic output in one CB-CD34+ sample and increased the same clonogenic output by 6.7-fold in the second CB sample, with no effect in LTC-IC output in CD34+ cells purified from PBSC. Amongst CML samples, the numbers of LTC-IC-derived progenitors were reduced 3-fold after shRNA-mediated STAT3 inhibition in one patient (UPN2). In all other 7 patients, inhibition of STAT3 by shRNA led to either stable ( n = 1, UPN 4) or increased ( n= 6 ) LTC-IC derived clonogenic activity, with major increase of 5-week clonogenic output in 3 patients (luciferase vs shSTAT3 clonogenic outputs 28 vs 353 for UPN5; 130 vs 270 for UPN6; 295 vs 806 for UPN8). Thus, our results suggest that STAT3 activation seen in primary CML leukemic cells does not play a role in stem cell self-renewal detectable by LTC-IC assays. On the contrary, STAT3 inhibition seems to lead to a stimulating effect of primitive stem cells in the majority of the patients analyzed. These findings do not rule out the potential role of STAT3 in BCR-ABL induced leukemogenesis but suggest that STAT3 inhibition is not a clinically useful target at the stem cell level in CML.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2647-2647 ◽  
Author(s):  
Linda Resar ◽  
Lingling Xian ◽  
Tait Huso ◽  
Amy Belton ◽  
Leslie Cope ◽  
...  

Abstract Introduction: Nuclear chromatin structure is a key determinant of stem cell function and cell fate, although factors that regulate this are only beginning to emerge. While High Mobility Group A1(HMGA1) chromatin remodeling proteins are among the most abundant, nonhistone chromatin binding proteins in adult stem cells (ASCs), their role in this setting has been unknown. HMGA1/2 proteins modulate gene expression by binding to DNA, bending chromatin, and recruiting transcription factor complexes to enhancers throughout the genome. The HMGA1 gene is highly expressed during embryogenesis with low or undetectable levels in mature, differentiated tissues. In cancer, HMGA1 re-expression occurs through oncogenic transcription factors, other epigenetic alterations, or in rare cases, chromosomal translocation events. Importantly, HMGA1 levels correlate with adverse clinical outcomes in diverse malignancies. We previously reported that Hmga1 transgenic mice develop leukemic transformation by inducing transcriptional networks involved in stem cell function and cell cycle progression. Methods: To elucidate the role of Hmga1 in normal development and ASCs in vivo, we generated mouse models with transgenic overexpression or deletion of Hmga1. To define the function of Hmga1 in adult stem cells (ASCs), we used gain-of-function (overexpression) and loss-of-function (silencing or genetic deletion) approaches in human and murine intestinal stem cells (ISCs) and hematopoietic stem and progenitor cells. Results:Transgenic mice overexpressing Hmga1 in ISCs develop hyperproliferation, aberrant crypt formation, and polyposis in the intestinal epithelium by expanding the ISC and niche compartments. Hmga1 enhances self-renewal in ISCs by amplifying Wnt/β-catenin signaling, inducing genes that encode both Wnt agonist receptors and downstream Wnt effectors. Surprisingly, Hmga1 also "builds" an epithelial niche by directly up-regulating Sox9 to induce Paneth cell differentiation. Paneth cells constitute the epithelial ISC niche by secreting Wnt agonists. This is the first example of Hmga1 fostering terminal differentiation to establish a stem cell niche. In human intestine, HMGA1 and SOX9 are highly correlated, and both become up-regulated in colorectal cancer. Human CD34+ cells engineered to overexpress Hmga1 expand more efficiently, while those with Hmga1 deficiency have defective proliferation and colony forming capability. Both colony number and size were decreased, and differentiation was skewed towards myeloid lineages. In mice, Hmga1 deletion causes partial embryonic lethality; over 50% of expected offspring die before mid-gestation. Those that survive develop premature aging phenotypes with early kyphosis, decreased bone density, grip strength, gait velocity, and hearing deficits. Knock-out mice also have early thymic aplasia, decreased numbers of early T-cell precursors, as well as decreased B-cell differentiation. Long-term (LT)-hematopoietic stem cells were decreased and preliminary data suggests aberrant regenerative function in serial, competitive transplant experiments.Preliminary ChIP-seq and gene expression studies in CD34+ cells suggest that Hmga1 regulates transcriptional networks involved in Wnt, JAK-STAT, and PI3K signaling. Conclusions:Our results in ASCs reveal a novel role for Hmga1 in tissue homeostasis by inducing pathways involved in Wnt and regenerative function. In ISCs, Hmga1 maintains both the stem cell pool and niche compartment whereas deregulated Hmga1 may perturb this equilibrium during carcinogenesis. Functional studies in HSCs suggest that Hmga1 also regulates self-renewal, regenerative potential, and the capacity for balanced differentiation. These findings indicate that HMGA1 is required for normal stem cell function, both during embryogenesis, and postnatally, in ASCs. Our prior work in tumor models demonstrates that a subset of HMGA1 stem cell pathways are hi-jacked by cancer cells to drive tumor progression. Together, these studies provide compelling rationale for further research to determine how to harness HMGA1 for regenerative medicine and to target it in cancer therapy. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 2017 ◽  
pp. 1-14 ◽  
Author(s):  
Ana Almazán-Moga ◽  
Patricia Zarzosa ◽  
Isaac Vidal ◽  
Carla Molist ◽  
Irina Giralt ◽  
...  

Rhabdomyosarcoma (RMS) is the most common type of soft tissue sarcoma in children and can be divided into two main subtypes: embryonal (eRMS) and alveolar (aRMS). Among the cellular heterogeneity of tumors, the existence of a small fraction of cells called cancer stem cells (CSC), thought to be responsible for the onset and propagation of cancer, has been demonstrated in some neoplasia. Although the existence of CSC has been reported for eRMS, their existence in aRMS, the most malignant subtype, has not been demonstrated to date. Given the lack of suitable markers to identify this subpopulation in aRMS, we used cancer stem cell-enriched supracellular structures (spheres and holoclones) to study this subpopulation. This strategy allowed us to demonstrate the capacity of both aRMS and eRMS cells to form these structures and retain self-renewal capacity. Furthermore, cells contained in spheres and holoclones showed significant Hedgehog pathway induction, the inhibition of which (pharmacologic or genetic) impairs the formation of both holoclones and spheres. Our findings point to a crucial role of this pathway in the maintenance of these structures and suggest that Hedgehog pathway targeting in CSC may have great potential in preventing local relapses and metastases.


2017 ◽  
Author(s):  
Patricia Rojas-Ríos ◽  
Aymeric Chartier ◽  
Martine Simonelig

AbstractPIWI proteins have essential roles in germ cells and stem cell lineages. In Drosophila, Piwi is required in somatic niche cells and germline stem cells (GSCs) for GSC self-renewal and differentiation. Whether and how other PIWI proteins are involved in GSC biology remains unknown. Here, we show that Aubergine (Aub), another PIWI protein, is intrinsically required in GSCs for their self-renewal and differentiation. Aub loading with piRNAs is essential for these functions. The major role of Aub is in self-renewal and depends on mRNA regulation. We identify the Cbl proto-oncogene, a regulator of mammalian hematopoietic stem cells, as a novel GSC differentiation factor. Aub represses Cbl mRNA translation for GSC self-renewal, and does so through recruitment of the CCR4-NOT complex. This study reveals the role of piRNAs and PIWI proteins in translational repression for stem cell homeostasis and highlights piRNAs as major post-transcriptional regulators in key developmental decisions.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5008-5008
Author(s):  
Lingyu Zeng ◽  
Wenyi Lu ◽  
Lan Ding ◽  
Wen Ju ◽  
Jianlin Qiao ◽  
...  

Introduction: Endothelial cells (ECs) provide a fertile niche for hematopoietic stem cell (HSC) maintenance, differentiation, and migration.Several studies have indicated that bone marrow (BM) vascular niche was impaired after HSC transplantation and severely inhibited hematopoietic reconstruction. Pigment epithelium-derived factor (PEDF) is an important potential cytoprotection and therapeutic agent for injured cells. The direct role of the injured endothelial cells on hematopoietic stem cells and whether PEDF has protective effect in this system remain unknown. This study aims to observe the influence of enjured ECs on HSCs and to explore the role of PEDF in endothelial-HSC coculture system. Methods: Injury of Endothelial cells by two important preparative regimenconditioning radiation and Busulfan respectively was evaluated with CCK8 assay. The expression of endothelial tight junctions(TJs),adherent junctions related molecules and endothelial to Mesenchymal Transition molecules such as ZO-1, Occludin,VE-cadherin, ICAM, α-SMA, CD31 and VCAM were detected by RT-qPCR, flow cytometry, immunofluorescence and western blot. The effects of injured endothelial cells on HSC self-renewal, differentiation, cell cycle and apoptosis were evaluated by flow cytometry, photography, viable cell count and clone formation assay. Hematopoiesis regulation factors SCF, IL-6, TGF-β and TNF-α were detected by ELISA. The protective effect of PEDF was also explored. Results: Both radiation and Busulfan could decrease cell viability of endothelial cells. The expression level of ZO-1, Occludin, VE-cadherin, ICAM, CD31 and VCAM were decreased and α-SMA was increased when EC exposed to radiation or Busulfan suggesting endothelial activation, impaired EC permeability and endothelial to Mesenchymal Transition after EC injured. Compared with normal endothelial cells and hematopoietic stem cell co-culture group, the HSC% of injured endothelial cells and hematopoietic stem cells co-cultured group were significantly decreased, the cell colony formation ability was decreased, the proportion of mature cells increased, and the damage of endothelial cells could not maintain the characteristics of HSC, weakened the self-renewal and multidirectional differentiation potential of HSC and promoted the maturation of HSC. After the administration of PEDF, endothelial to Mesenchymal Transition of EC was suppressed and the EC permeability was improved. Most importantly, the proportion of HSC was significantly increased, and the proportion of mature cells decreased in the coculture system. Conclusion: Injured endothelial cells can inhibit proliferation of hematopoietic stem cells, self-renewal and promote HSC differentiation. PEDF could ameliorate endothelial injury and promote HSC expansion by suppressing endothelial-mesenchymal transition and protecting TJs and AJs. Disclosures No relevant conflicts of interest to declare.


2015 ◽  
Vol 27 (1) ◽  
pp. 258
Author(s):  
M. Duque ◽  
M. N. Biancardi ◽  
J. H. Galiguis ◽  
C. E. Pope ◽  
C. Dumas ◽  
...  

Different feeder cells (FC) influence the isolation, proliferation, and self-renewal of cat embryonic stem cells (cat ESC; Gómez et al. 2010 Theriogenology 74, 498–515) possibly by secretion of growth factors that affect intracellular signalling pathways involved in self-renewal. Supplementation of the culture medium with fibroblast growth factor (FGF) stimulates the secretion of Activin A in mouse and human FC, which enhances undifferentiation in human ESC (Eiselleova et al. 2008 Int. J. Dev. Biol. 52, 353-363). Moreover, the Activin/Nodal pathway plays an important role in maintaining pluripotency of hESC through mechanism(s) in which FGF acts as a competence factor (Vallier et al. 2005 J. Cell Sci. 118, 4495–4509). Little is known about secretion of growth factors by cat FC and whether cat ESC use the activin/nodal pathway for their self-renewal. Our previous work has indicated that culturing cat ESC with bFGF enhances the stem cell replication and self-renewal (Gómez et al. 2010 Theriogenology 74, 498–515). Here we evaluated the effect of bFGF supplementation in the culture medium on the abilities of cat embryonic fibroblast (CEF) and mouse embryonic fibroblast (MEF) FC to: (1) secrete Activin A and (2) support undifferentiated growth of cat ESC. For experiment 1, mitomycin-C-treated CEF (n = 2) and MEF (n = 2) were, respectively, cultured with ESC medium supplemented with (1) LIF (1000 IU), (2) bFGF (10 ng mL–1), (3) LIF + bFGF, or (4) no factors. The medium for each condition was collected at 24 h after culture and Activin A protein concentration was detected with a feline Activin A-ELISA kit. Results showed that supplementation of ESC medium with bFGF with or without LIF significantly increased the secretion of Activin A in MEF (5256 and 7048 ng mL–1, respectively; P < 0.001), but not in CEF (150 and 131 ng mL–1, respectively). Moreover, differences in Activin A secretion were observed between both MEF cell lines (10 269 v. 2034 ng mL–1; P < 0.001). For experiment 2, cat ESC were cultured in CEF or MEF in the ESC medium supplemented with bFGF (10 ng mL–1), LIF (1000 UI), and an inhibitor of glycogen synthase kinase-3 β (GSK3-b), SB 216763 (2.1 µM mL–1). Results showed differences in morphology of cat ESC cultured in CEF or MEF, where colonies cultured in CEF had clearly defined borders and a tightly domed shape, with a high nucleus to cytoplasm ratio and prominent nucleoli. In comparison, ESC cultured in MEF had poorly defined borders and a flattened shape. In addition, the mean cell size of colonies at passage 8 (P8) cultured on CEF was larger (612 ± 0.9 µm) than that of those cultured on MEF (360 ± 0.5 µm; P < 0.001). Colonies cultured on MEF differentiated into fibroblast-like cells and other noncharacterised cell types after P8. These results clearly indicated that CEF do not secrete Activin A. The negative effect of Activin A on the morphology of cat ESC cultured on MEF may suggest a synergism between GSK3b inhibitor and Activin A that may induce differentiation, possibly into mesoendodermal cells (Teo et al. 2014 Stem Cell Rep. 3, 5–14). Studies that evaluate the effects of supplementing ESC medium with a lower concentration of Activin A may help to elucidate the importance of the Activin/Nodal pathway in cat ESC.


2021 ◽  
Vol 11 (9) ◽  
pp. 1780-1784
Author(s):  
Kewei Chen ◽  
Yiqin Ouyang ◽  
Junhua Liang ◽  
Huaifang Li ◽  
Xiaowen Tong ◽  
...  

We aimed to explore the mechanism underlying the role of miR-21 derived from bone marrow mesenchymal stem cell exosomes (BMSC-exos) in cervical cancer (CC) and the relation between angiogenesis and autophagy. In this study, BMSC-exos were co-cultured with CC stem cells followed by analysis of miR-21 expression by RT-qPCR, autophagy after hunger-induced feeding by Acridine Orange fluorescent staining, angiogenesis by tube formation assay. Co-culture of BMSC-exos effectively reduced miR-21 expression in CC stem cells and enhanced autophagy as demonstrated by upregulated Beclin1 and LC3B with assembly of autophagosome (p < 0.05), but the autophagy restored later. Moreover, in the presence of BMSC-exos, CC stem cell angiogenesis was suppressed by 79%. In conclusion, BMSC-exos enhance autophagy and inhibit angiogenesis in CC through decreasing miR-21, which provides a novel insight into etiology of CC.


Sign in / Sign up

Export Citation Format

Share Document