scholarly journals Pentosan polysulfate binds to STRO-1+ mesenchymal progenitor cells, is internalized, and modifies gene expression: a novel approach of pre-programing stem cells for therapeutic application requiring their chondrogenesis

2017 ◽  
Vol 8 (1) ◽  
Author(s):  
Jiehua Wu ◽  
Susan Shimmon ◽  
Sharon Paton ◽  
Christopher Daly ◽  
Tony Goldschlager ◽  
...  
Circulation ◽  
2008 ◽  
Vol 118 (suppl_18) ◽  
Author(s):  
Zhilin Chen ◽  
Sean R Hall ◽  
Keith R Brunt ◽  
Zhan Liu ◽  
David A Ademidun ◽  
...  

Human stem and progenitor cells have emerged as potentially useful substrates for cardiovascular repair through neovascularization and myocardial regeneration. However, efficacy is limited by impedance to stem cell retention, homing and differentiation in hostile microenvironments, as occur in infarcted myocardium. The objective of the current study was to regulate gene function for tailored therapy in post infarct myocardium. Here we show that hypoxic and inflammatory stimuli of the infarct microenvironment regulate a proportional response in gene expression in human endothelial progenitor (EPC) and mesenchymal stem cells (MSC). Highly efficient lentiviral vectors incorporating hypoxia (HRE) and nuclear factor kappa B (NFkB) responsive elements are used to drive transgenes for survival, autologous stem cell homing and cardiogenic differentiation. Utilizing an internal cytomegalovirus promoter deleted lentiviral transfer vector, an HRE-NFkB bicistronic promoter-reporter vector was constructed with a modified internal ribosome entry sequence between green fluorescent protein and luciferase or therapeutic genes. Either hypoxia or inflammation resulted in a seven to ten-fold response of transgene expression assessed by luciferase activity in EPC (hypoxia, 7608±954; inflammation 11492±1384, P<0.01 and P<0.001 vs control 1049±139 respectively, N=6), while combined hypoxic-inflammatory stimuli resulted in a sixty-fold increase of transgene expression (hypoxic-inflammation, 62364±6609, P<0.001 vs control 1049±139, N=6). These results were recapitulated in MSC and with a series of therapeutic genes as determined by transcript, protein expression and activity. Our results demonstrate that regulated vectors provide a proportional response to hostile post-infarct myocardium. Translating cardiovascular regenerative medicine using stem cells requires managing stem cell survival, function and differentiation. Utilizing site-specific pathophysiological cues to auto-regulate reparative and regenerative gene expression, this study is a starting point for sophisticated platforms for patient tailored cell-based cardiogenic therapy.


2002 ◽  
Vol 20 (1) ◽  
pp. 110-123 ◽  
Author(s):  
Vincent F. La Russa ◽  
Paul Schwarzenberger ◽  
Alan Miller ◽  
Krishna Agrawal ◽  
Jay Kolls ◽  
...  

QJM ◽  
2020 ◽  
Vol 113 (Supplement_1) ◽  
Author(s):  
D Sabry ◽  
W A Khalifa ◽  
M M Abdelgwad ◽  
M Alhelf ◽  
Z M Altaib

Abstract Background Bone marrow mesenchymal stem cells (BM-MSCs) and human umbilical cord endothelial progenitor cells (UC-EPCs) have several benefits for liver regeneration. We speculate huge impacts of rat BM-MSCs and UC-EPCs on reversal of hepatic injury and improvement of liver function in liver fibrosis induced by carbon tetrachloride (CCl4). Methods Forty adult rats were divided into 4 groups; control group, CCl4 group, CCl4/BM-MSCs group and CCl4/UCEPCs group. Blood samples were driven from rats at 1, 2 and 3months to measure serum concentration of albumin and alanine aminotransferase (ALT). Quantitative expression of HGF,TGF-β, MMP-2, and VEGF were assessed by polymerase chain reaction. Histological examination of the liver tissue was performed. α-SMA immunohistochemistry to identify the myoepithelial cells (MECs) and Ki-67 to identify cell prolifration immunohistochemistry are detected in groups injected with cells to confirm cells regeneration. Results Regarding liver function, there was elevating albumin (P &lt; 0.05) and reducing ALT (P &lt; 0.05) concentrations in groups treated with BM-MSCs and UC-EPCs effect compared to untreated CCL4 group. Concerning gene expression, UC-EPCs treated group have significantly higher MMP-2 and VEGF (P &lt; 0.01) genes expression than BM-MSCs treated group. Furthermore, UC-EPCs were more valuable than BM-MSCs in increasing gene expression of HGF (P &lt; 0.05) and immunohistochemistry of α-SMA and Ki-67 (P &lt; 0.01). BM-MSCs have significantly lower TGF- β (P &lt; 0.00) compared to UC-EPCs. Conclusion This study highlighted on liver regeneration role of both human UC-EPCs and BM-MSCs in liver fibrosis by different signaling mechanistic.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4105-4105
Author(s):  
Brenton Short ◽  
Emer Clarke ◽  
Terry Thomas ◽  
Allen C. Eaves ◽  
Albertus W. Wognum ◽  
...  

Abstract Adult mammalian bone marrow (BM) contains at least two distinct stem cell populations; the stem cells of the hemopoietic lineage and a second population termed mesenchymal stem cells (MSC) whose function is to maintain the non-hemopoietic BM elements as well as skeletal homeostasis. MSC have been implicated as potential targets in a range of cellular therapies for treatment of defects of both the hemopoietic and skeletal systems, and as vehicles for gene therapy. In order to evaluate the potential of these cells in various therapies, a pre-clinical animal model in which both the biology and potential therapeutic applications of these cells can be assessed is of fundamental importance. The goal of the current study was to develop a robust and reproducible method for the isolation of MSC from murine hemopoietic tissues. Tibiae and femurs harvested from C57BL6/J mice were gently crushed with a pestle to release the marrow. The bone fragments were subsequently cut into small pieces with a scalpel and digested in a solution containing 3mg/ml Type I collagenase to yield a population of compact bone (CB) derived cells. Mesenchymal progenitor cells (MPC) were detected using an in vitro assay for fibroblast-colony forming cells (CFU-F). CB cells were plated at 1000 or 5000 cells per cm2 in complete MesenCult™ medium for 12 days, after which CFU-F-derived colonies were enumerated. We show that CFU-F are present at a significantly higher frequency in mouse CB than in the BM (433±225 vs 11.7±3.5 colonies/106 cells respectively, n=3). Based on these data we developed a simple and robust immunomagnetic selection method to highly enrich MPC from mouse CB by depleting essentially all nucleated hemopoietic cells (CD45+) and red blood cells (Ter119+) using magnetic particles and antibodies to CD45 and Ter119, respectively. Target CD45−Ter119− cells initially comprised 1.1±0.5% (n=9) of the total CB fraction as assayed by FACS. Following depletion, CD45−Ter119− cells comprised 74.5±16% (n=6) of the cells and were enriched 205 fold for CFU-F compared to the starting population, with a CFU-F frequency of 1 per 11 cells plated, and a total CFU-F recovery of 57.9 ± 18.5%. Analysis of CD45−Ter119−Sca-1+ cells, a phenotype previously shown to enrich for MPC, revealed that these cells were enriched 50 fold following depletion, from 0.53±0.5 to 26.5±8.23% (n=3). The enriched MPCs cultured at low O2 tension were devoid of hemopoietic contaminants at passage 1 and 2 as shown by lack of CD45, Ter119 and CD11b expression. The cultured CB-derived MPCs were capable of extensive in vitro proliferation and maintained the ability to differentiate into cells of the osteogenic, adipogenic and chondrogenic lineages. Furthermore, irradiated cultured mesenchymal cells supported long-term culture-initiating cells (LTC-IC) in 4-week cultures of Sca-1+ BM cells under limiting dilution conditions, at frequencies similar to those detected using irradiated primary BM feeders (i.e. 1 per 1600). These data provide a rapid, reproducible method by which multipotent mesenchymal cells devoid of contaminating hemopoietic cells can be readily obtained from limited numbers of mice to study the biology of MSC as well as the use of these cells as therapeutic agents in a preclinical animal model.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2588-2588 ◽  
Author(s):  
Louise Roulin ◽  
Ashfaq Ali ◽  
Aline Masse ◽  
Marie-Magdelaine Coudé ◽  
Dominique Bluteau ◽  
...  

Abstract CONTEXT: Eradication of leukemic progenitor cells, defined by functional assays such as long-term culture (leukemic long-term culture initiating cells [L-LTC-IC]) is the goal of therapy in AML. Bromodomain and ExtraTerminal (BET) proteins are epigenetic readers that regulate the expression of genes with super-enhancers, including CMYC. BET inhibitors (BETi) such as JQ1 induce proliferation arrest and apoptosis in murine models of AML, in human AML cell lines and primary blasts. Their activity in human leukemic progenitors has not yet been reported. OTX015 (MK-8626) is an orally available BETi that can be safely administered to patients with a continuous low-dose regimen (Dombret et al. Blood. 2014). Single-dose exposure to OTX015 induces gene expression modulation characteristic of bromodomain inhibition, including downregulation of CMYC and upregulation of HEXIM1, inhibiting the viability of AML cell lines, and inducing apoptosis in primary AML blasts (Coudé et al. Oncotarget. 2015). To address the activity of OTX015 on leukemic progenitors, we analyzed (A) the clonogenicity of AML cell lines and (B) the frequency of primary L-LTC-IC after repeated low-dose exposure to OTX015. METHODS: (A) Five AML cell lines (OTX015 IC50 60 - 10,000 nM) were studied: OCI-AML3, NOMO-1, HL-60, KG1a and K562. After 24h starvation, OTX015 or vehicle (DMSO) was added daily to the culture medium for 3 days at various concentrations. After 96h, cells were assessed for gene expression by RT-qPCR and seeded in methycellulose. Colonies were scored after 14 days. (B) Bone-marrow mononuclear cells (BMNC) from AML patients obtained at diagnosis after informed consent were cultured for three weeks in a niche-like hypoxic milieu shown to maintain leukemic stem cells (Griessinger et al. Stem Cells Transl Med. 2014). OTX015 200 nM or DMSO was added weekly. This concentration is in the range of trough concentrations achievable at the MTD of OTX015 in phase I trials. Residual leukemic cells were sorted and plated on methylcellulose. Colonies were scored after 14 days. The resulting L-LTC-IC frequency was reported relative to the number of BMNC initially seeded. RESULTS: (A) To dissect the effect of OTX015 on AML progenitors from that on the leukemic bulk, we determined for each cell line a maximal OTX015 concentration that could be administered repeatedly for 3 days without significantly impairing proliferation or viability (MTT) at day 4 of culture (referred as low-dose concentration). As expected, this target concentration, ranging from 50 to 500 nM, was lower in cell lines with low OTX015 IC50. This prolonged low-dose exposure to OTX015 recapitulated BETi-associated gene expression changes including CMYC downregulation and HEXIM1 upregulation in all cell lines, and significantly reduced clonogenicity compared to DMSO in 4/5 cell lines, but not in NPM1-mutated OCI-AML3 cells (IC50: 60 nM, target concentration 50 nM), despite modulation of CMYC and HEXIM1 expression. Overall, there was no correlation between the level of CMYC repression and clonogenicity. Transcriptome analyses are ongoing to identify gene expression changes specifically associated with inhibition of clonogenicity. (B) L-LTC-IC frequency after prolonged exposure to 200 nM OTX015 was determined in specimens from 11 AML patients with variable oncogenetics. L-LTC-IC frequency was reduced in 5/11 patients, reaching statistical significance in 3 cases; OTX015 reduced L-L-LTC-IC in 3 of 4 NPM1-mutated samples, but not in any of the 3 patients with high-risk cytogenetics. No clear correlation was found between induction of apoptosis on primary blasts after short-term, and L-LTC-IC reduction after long-term 200nM OTX015 exposure respectively. Patients' samples number is being extended to identify oncogenetic predictors of L-LTC-IC reduction. CONCLUSION: Our results suggest that in AML cell lines or primary samples, prolonged exposure to low concentrations of the clinically-available BET inhibitor OTX015 results in activity against leukemic progenitors independent of induction of proliferation arrest or apoptosis in blasts. Molecular mechanisms and oncogenic markers of this activity are being investigated. These results warrant clinical investigation of the anti-leukemic properties of prolonged low-dose OTX015 administration. Disclosures Riveiro: Oncoethix: Research Funding; OTD: Employment. Herait:Oncoethix: Other: shareholder; Oncoethix: Other: Chief medical officer; Oncoethix: Other: shareholder. Dombret:Oncoethix: Research Funding. Itzykson:Oncoethix: Research Funding.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2232-2232
Author(s):  
Mario Schubert ◽  
Margot Zoeller ◽  
Wolfgang Wagner ◽  
Patrick Wuchter ◽  
Volker Eckstein ◽  
...  

Abstract Introduction. We have shown in the past that slow divisional kinetics of hematopoietic stem cells (HSC) are correlated with prolonged self renewal and increased frequency of long term culture initiating cells (LTC-IC) and myeloid-lymphoid initiating cells (ML-IC) in vitro. In the present study we have demonstrated that only the slow dividing fraction (SDF) of human CD34+ HSC, but not the fast dividing fraction (FDF), is able to engraft in the thymus and lymph nodes of NOD/SCID mice. Methods. Human CD34+ HSC were isolated from umbilical cord blood, stained with the membrane fluorescence marker PKH26, cultivated in cytokine rich media for five days and then sorted for their PKH26 fluorescence (dim versus bright). Sorted cells were injected into the tail vein of sublethally irradiated NOD/SCID mice (2×10e5 cells / animal). Mice were sacrificed at defined time points from week 2 to week 18 after transplantation. Results. Flow cytometric analysis of the samples collected from the marrow, spleen, thymus, and lymph nodes of the NOD/SCID mice indicated that both SDF and FDF were able to engraft in the bone marrow of the mice, establishing a hematopoietic system, i.e. erythroid and myeloid progenitor cells were recovered from the bone marrow of SDF and FDF reconstituted mice. The most remarkable finding was that only progeny cells of SDF were found in the mouse lymph nodes and thymus where, beginning approximately at week 15, they differentiated into T cells. CD4+CD8+ double positive T progenitor cells were recovered in the thymus and CD4+ as well as CD8+ T cells were present in the spleen and the lymph nodes. However, at least until 18 weeks after reconstitution, very few B cells were present in the NOD/SCID spleen, whereas monocytes reached close to normal values. We are in the process of evaluating the functionality of the human T cells that matured in the NOD/SCID mouse environment. Global gene expression profiles of the two subpopulations (SDF and FDF) were analyzed using a ‘human transcriptome cDNA microarray’. We found that several molecular markers for stem cells were highly expressed in the SDF as compared to FDF: CD133 (Prominin), MDR1 (multiple drug resistance gene 1), clqr1 (complement component 1 receptor 1), Hoxa9, Cdx1 and Hesx1 (which encode homeodomain proteins). Conclusion. Our data indicated that only SDF was enriched for primitive HSC, which in turn were able to engraft in the thymus and lymph nodes in the SCID mouse transplantation model. These cells were able to give rise to T cell precursors and mature T cells. This observation has provided unequivocal evidence that the SDF is associated with primitive HSC function, and is consistent with the results of differential gene expression analysis in SDF versus FDF. The significance and nature of the cell-cell contacts in the microenvironment of the SCID mouse model is currently being examined.


2018 ◽  
Vol 19 (9) ◽  
pp. 2549 ◽  
Author(s):  
Luisa Brandt ◽  
Susanna Schubert ◽  
Patrick Scheibe ◽  
Walter Brehm ◽  
Jan Franzen ◽  
...  

Transplantation of multipotent mesenchymal progenitor cells is a valuable option for treating tendon disease. Tenogenic differentiation leading to cell replacement and subsequent matrix modulation may contribute to the regenerative effects of these cells, but it is unclear whether this occurs in the inflammatory environment of acute tendon disease. Equine adipose-derived stromal cells (ASC) were cultured as monolayers or on decellularized tendon scaffolds in static or dynamic conditions, the latter represented by cyclic stretching. The impact of different inflammatory conditions, as represented by supplementation with interleukin-1β and/or tumor necrosis factor-α or by co-culture with allogeneic peripheral blood leukocytes, on ASC functional properties was investigated. High cytokine concentrations increased ASC proliferation and osteogenic differentiation, but decreased chondrogenic differentiation and ASC viability in scaffold culture, as well as tendon scaffold repopulation, and strongly influenced musculoskeletal gene expression. Effects regarding the latter differed between the monolayer and scaffold cultures. Leukocytes rather decreased ASC proliferation, but had similar effects on viability and musculoskeletal gene expression. This included decreased expression of the tenogenic transcription factor scleraxis by an inflammatory environment throughout culture conditions. The data demonstrate that ASC tenogenic properties are compromised in an inflammatory environment, with relevance to their possible mechanisms of action in acute tendon disease.


Sign in / Sign up

Export Citation Format

Share Document