scholarly journals Effect of bone marrow-derived mesenchymal stem cells and stem cell supernatant on equine corneal wound healing in vitro

2017 ◽  
Vol 8 (1) ◽  
Author(s):  
Amanda B. Sherman ◽  
Brian C. Gilger ◽  
Alix K. Berglund ◽  
Lauren V. Schnabel
2019 ◽  
Vol 2019 ◽  
pp. 1-16 ◽  
Author(s):  
Xudong Wang ◽  
Tongzhou Liang ◽  
Jincheng Qiu ◽  
Xianjian Qiu ◽  
Bo Gao ◽  
...  

Mesenchymal stem cells (MSCs) are promising candidates for tissue regeneration and disease treatment. However, long-term in vitro culture results in loss of MSC stemness. The inflammation that occurs at stem cell transplant sites (such as that resulting from TNF-α) is a contributing factor for stem cell treatment failure. Currently, there is little evidence regarding the protective role of melatonin with regard to the negative effects of TNF-α on the stemness of MSCs. In this study, we report a melatonin-based method to reduce the inflammatory effects on the stemness of bone marrow mesenchymal stem cells (BMMSCs). The results of colony formation assays, Alizarin red staining, western blotting, and reverse transcription-polymerase chain reactions suggest that melatonin can reverse the inflammatory damage caused by TNF-α treatment in the third, seventh, and tenth generations of primary BMMSCs (vs. control and the TNF-α-treated group). Meanwhile, a detailed analysis of the molecular mechanisms showed that the melatonin receptor and YAP signaling pathway are closely related to the role that melatonin plays in negative inflammatory effects against BMMSCs. In addition, in vivo experiments showed that melatonin could reverse the damage caused by TNF-α on bone regeneration by BMMSCs in nude mice. Overall, our results suggest that melatonin can reverse the loss of stemness caused by inflammatory factor TNF-α in BMMSCs. Our results also provide a practical strategy for the application of BMMSCs in tissue engineering and cell therapy.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Ling Guo ◽  
Juan Du ◽  
Dan-feng Yuan ◽  
Ya Zhang ◽  
Shu Zhang ◽  
...  

Abstract Background The transplantation of bone marrow mesenchymal stem cells (BMSCs) is a promising therapeutic strategy for wound healing. However, the poor migration capacity and low survival rate of transplanted BMSCs in wounds weaken their potential application. Objective To identify the optimal protocol for BMSCs preconditioned with H2O2 and improve the therapeutic efficacy using H2O2-preconditioned BMSCs in wound healing. Methods Mouse BMSCs were exposed to various concentrations of H2O2, and the key cellular functional properties were assessed to determine the optimal precondition with H2O2. The H2O2-preconditioned BMSCs were transplanted into mice with full-thickness excisional wounds to evaluate their healing capacity and tissue engraftment. Results Treatment BMSCs with 50 μM H2O2 for 12 h could significantly enhance their proliferation, migration, and survival by maximizing the upregulation of cyclin D1, SDF-1, and its receptors CXCR4/7 expressions, and activating the PI3K/Akt/mTOR pathway, but inhibiting the expression of p16 and GSK-3β. Meanwhile, oxidative stress-induced BMSC apoptosis was also significantly attenuated by the same protocol pretreatment with a decreased ratio of Bax/Bcl-2 and cleaved caspase-9/3 expression. Moreover, after the identification of the optimal protocol of H2O2 precondition in vitro, the migration and tissue engraftment of transfused BMSCs with H2O2 preconditioning were dramatically increased into the wound site as compared to the un-preconditioned BMSCs. The increased microvessel density and the speedy closure of the wounds were observed after the transfusion of H2O2-preconditioned BMSCs. Conclusions The findings suggested that 50 μM H2O2 pretreated for 12 h is the optimal precondition for the transplantation of BMSCs, which gives a considerable insight that this protocol may be served as a promising candidate for improving the therapeutic potential of BMSCs for wound healing.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5976-5976
Author(s):  
Hwan-Young Kim ◽  
Ji-Eun Noh ◽  
Hee-Jo Baek ◽  
Jae-Dong Moon ◽  
Jong-Hee Shin ◽  
...  

Abstract Background: Fluoranthene is a member of the polycyclic aromatic hydrocarbon family, comprising ubiquitous environmental pollutants and potent mutagens. Fluoranthene has been classified by the International Agency for Research on Cancer as a group 3 carcinogens, whose carcinogenicity has not been fully determined in humans. Moreover, the stem cell toxicity and global hematotoxicity associated with fluoranthene exposure have not been thoroughly studied in the bone marrow-mesenchymal stem cells (BM-MSCs). In this study, we determined whether fluoranthene-induced cellular responses could be used as biomarkers for the examination of BM-MSC dysfunction, and suggested the possible use of BM-MSCs for monitoring the acute hematotoxicity caused by environmental hazards. Materials and Methods: Apreviously published protocol was used for the isolation and characterization of BM-MSCs. Fluoranthene was added to the cell culture medium in the range of 25–500 µM. The cellular levels of hydrogen peroxide, indicating the presence of reactive oxygen species, were measured using an enzyme immunoassay. The mitochondrial mass, membrane potential, and mitochondrial DNA (mtDNA) copy number were measured using MitoTracker Green, MitoTracker Red probes, and real time PCR, respectively. A proteomic analysis of the mitochondrial-rich cytoplasmic fraction was performed using nano-LC-ESI-MS/MS, BioWorksBrowser, and the SEQUEST search engines. Quantitative mRNA and immunoblot measurements were used to further confirm the altered mRNA expression as well as to determine the levels of cellular proteins obtained from the proteomic analysis. Results: After exposure to fluoranthene, the BM-MSCs showed a marked reduction in cell number, and the viability decreased substantially after two days of exposure. BM-MSCs that were not treated with fluoranthene remained compact and spindle-shaped. These cells remained tightly attached to each other and to the substrate. In general, a direct exposure of fluoranthene depressed the proliferative capacity and altered the cell morphology of BM-MSCs. The cells detached from the subsurface, and cell-to-cell attachments were also lost. The viability significantly decreased after two days of fluoranthene exposure. The mtDNA copy number and the mass showed a rapid elevation after a 5-day exposure to fluoranthene. Hundreds of cellular proteins in the mitochondria-rich cytoplasmic fraction were markedly deregulated in cells treated with fluoranthene. The protein expression levels of poly [ADP-ribose] polymerase 1 (PARP-1), elongation factor 1-gamma, heat shock 70 kDa protein 1A/1B, heterogeneous nuclear ribonucleoproteins A2/B1 isoform B1, ATP-dependent RNA helicase DDX5, and T-complex protein 1 subunit theta were upregulated more than five-fold in cells treated with fluoranthene than in untreated cells. A significant (more than 2-fold) down-regulation in the cellular levels of the proteins myosin-9, protein ALO17 isoform 1, filamin-C isoform b, Na/K-transporting ATPase subunit alpha-1, nuclear pore membrane glycoprotein 210, and DNA-dependent protein kinase catalytic subunit isoform 2 was observed after a similar fluoranthene treatment. The presence of PARP-1 was further confirmed using mRNA analysis. Conclusion: This study investigated the global cellular responses after exposure to fluoranthene: PARP-1 was recognized as a notable biomarker for monitoring the PAH-induced hematotoxicity. In summary, BM-MSCs are promising candidates for the development of unique in vitro model systems for predicting fluoranthene-associated hematotoxicity and general toxicity in humans. Keywords: Fluoranthene, genotoxicity, hematotoxicity, bone marrow-mesenchymal stem cells Figure 1. Stem cell-based in vitro models and biomarkers for studying the hematotoxic effects of fluoranthene exposure. (A) Direct exposure of PAHs depressed the proliferative capacity of h-TERT cells with a thread-like or round shape and loose cell-to-cell attachment. (B) Cytotoxic effect of fluoranthene exposure to the bone marrow-mesenchymal stem cells remarkably increased with dose-dependent manner. (C) Identified potential biomarkers were categorized as their biological processes and molecular functions. (D) Immunoblot confirmed the increased expression of poly [ADP-ribose] polymerase 1 (PARP-1) after exposure of fluoranthene. Figure 1. Stem cell-based in vitro models and biomarkers for studying the hematotoxic effects of fluoranthene exposure. (A) Direct exposure of PAHs depressed the proliferative capacity of h-TERT cells with a thread-like or round shape and loose cell-to-cell attachment. (B) Cytotoxic effect of fluoranthene exposure to the bone marrow-mesenchymal stem cells remarkably increased with dose-dependent manner. (C) Identified potential biomarkers were categorized as their biological processes and molecular functions. (D) Immunoblot confirmed the increased expression of poly [ADP-ribose] polymerase 1 (PARP-1) after exposure of fluoranthene. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 2019 ◽  
pp. 1-12 ◽  
Author(s):  
Jianing Ding ◽  
Xin Wang ◽  
Bi Chen ◽  
Jieyuan Zhang ◽  
Jianguang Xu

The exosomes are derived from mesenchymal stem cells (MSCs) and may be potentially used as an alternative for cell therapy, for treating diabetic wounds, and aid in angiogenesis. This study, aimed to investigate whether exosomes originated from bone marrow-derived MSCs (BMSCs) preconditioned by deferoxamine (DFO-Exos) exhibited superior proangiogenic property in wound repair and to explore the underlying mechanisms involved. Human umbilical vein endothelial cells (HUVECs) were used for assays involving cell proliferation, scratch wound healing, and tube formation. To test the effects in vivo, streptozotocin-induced diabetic rats were established. Two weeks after the procedure, histological analysis was used to measure wound-healing effects, and the neovascularization was evaluated as well. Our findings demonstrated that DFO-Exos activate the PI3K/AKT signaling pathway via miR-126 mediated PTEN downregulation to stimulate angiogenesis in vitro. This contributed to enhanced wound healing and angiogenesis in streptozotocin-induced diabetic rats in vivo. Our results suggest that, in cell-free therapies, exosomes derived from DFO preconditioned stem cells manifest increased proangiogenic ability.


2012 ◽  
Vol 90 (5) ◽  
pp. e367-e373 ◽  
Author(s):  
Saadettin Sel ◽  
Ulf M. Schilling ◽  
Norbert Nass ◽  
Andreas Simm ◽  
Fabian Garreis ◽  
...  

2010 ◽  
Vol 22 (1) ◽  
pp. 351
Author(s):  
A. J. Maki ◽  
I. Omelogu ◽  
E. Monaco ◽  
M. E. McGee-Lawrence ◽  
R. M. Bradford ◽  
...  

During winter hibernation, grizzly bears (Ursus arctos horribilis) do not eat but instead rely on internal fat stores as a primary source of metabolic energy. The resulting seasonal fluctuations in appetite and body mass make the grizzly bear a naturally occurring animal model for human conditions such as obesity and anorexia. An in vitro model of hibernating bear stem cells might enhance our understanding of processes such as stem cell proliferation and differentiation. Mesenchymal stem cells, derived from bone marrow and adipose tissue among others, differentiate into adipocytes and might play important roles in energy metabolism. In the current study, we examined the in vitro viability and morphology of mesenchymal stem cells isolated from grizzly bear adipose tissue (ADSC) and bone marrow (BMSC); these ADSC and BMSCs underwent adipogenic differentiation for 0, 7, 14, 21, and 28 days. Bone marrow stem cells and ADSC were isolated using mechanical disaggregation, collagenase digestion, centrifugation, and plating onto tissue culture polystyrene. Cell viability and proliferation was quantified using the colony forming unit assay and a hemocytometer. Both stem cell types were differentiated into adipocytes using 10 μM insulin, 1 μM dexamethasone, and 0.5 mM isobutylmethylxanthine (all Sigma- Aldrich, St. Louis, MO, USA) with the addition of 10% fetal bovine (FBS) or bear serum from the active feeding period. Adipogenic differentiation was confirmed using Oil Red O and quantified using ImageJ. Statistical analysis was performed using an unpaired t-test between treatments of the same time point. All cells were isolated within 28 h of tissue harvest. Adipose-derived stem cells formed an average of 11 colonies (0.011%), whereas BMSC formed 1.5 colonies (0.0015%) per 100 000 cells. Doubling time forADSC was approximately 54 h in 10% FBS. BothADSC and BMSC had an initial spindle-shaped morphology, which gradually became more rounded during adipogenic differentiation. For bear serum at Day 28, ADSC had a significantly (P < 0.01) greater stained area per cell than did BMSC. In summary, both types of mesenchymal stem cells successfully differentiated into adipocytes and maintained viability. In conclusion, grizzly bear mesenchymal stem cells canbesuccessfully isolated, expanded, and differentiated in culture. These results allow for future studies using the bear as an in vitro model for fat metabolism during hibernation and active periods. This work was partially supported by the Carle Foundation Hospital, the Intel Scholar’s Research Program, USDA Multi-State Research Project W1171, and the Illinois Regenerative Medicine Institute (IDPH # 63080017). In addition, the authors would like to thank Agatha Luszpak for support with the analysis.


2018 ◽  
Vol 315 (6) ◽  
pp. C885-C896 ◽  
Author(s):  
Jianming Guo ◽  
Haidi Hu ◽  
Jolanta Gorecka ◽  
Hualong Bai ◽  
Hao He ◽  
...  

We have previously shown that bone marrow-derived mesenchymal stem cells (BMSC) accelerate wound healing in a diabetic mouse model. In this study, we hypothesized that adipose tissue-derived stem cells (ADSC), cells of greater translational potential to human therapy, improve diabetic wound healing to a similar extent as BMSC. In vitro, the characterization and function of murine ADSC and BMSC as well as human diabetic and nondiabetic ADSC were evaluated by flow cytometry, cell viability, and VEGF expression. In vivo, biomimetic collagen scaffolds containing murine ADSC or BMSC were used to treat splinted full-thickness excisional back wounds on diabetic C57BL/6 mice, and human healthy and diabetic ADSC were used to treat back wounds on nude mice. Wound healing was evaluated by wound area, local VEGF-A expression, and count of CD31-positive cells. Delivery of murine ADSC or BMSC accelerated wound healing in diabetic mice to a similar extent, compared with acellular controls ( P < 0.0001). Histological analysis showed similarly increased cellular proliferation ( P < 0.0001), VEGF-A expression ( P = 0.0002), endothelial cell density ( P < 0.0001), numbers of macrophages ( P < 0.0001), and smooth muscle cells ( P < 0.0001) with ADSC and BMSC treatment, compared with controls. Cell survival and migration of ADSC and BMSC within the scaffolds were similar ( P = 0.781). Notch signaling was upregulated to a similar degree by both ADSC and BMSC. Diabetic and nondiabetic human ADSC expressed similar levels of VEGF-A ( P = 0.836) in vitro, as well as in scaffolds ( P = 1.000). Delivery of human diabetic and nondiabetic ADSC enhanced wound healing to a similar extent in a nude mouse wound model. Murine ADSC and BMSC delivered in a biomimetic-collagen scaffold are equivalent at enhancing diabetic wound healing. Human diabetic ADSC are not inferior to nondiabetic ADSC at accelerating wound healing in a nude mouse model. This data suggests that ADSC are a reasonable choice to evaluate for translational therapy in the treatment of human diabetic wounds.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2-2 ◽  
Author(s):  
Simón Méndez-Ferrer ◽  
Tatyana V. Michurina ◽  
Francesca Ferraro ◽  
Amin Mazloom ◽  
Ben MacArthur ◽  
...  

Abstract Abstract 2 Despite their therapeutic potential, mesenchymal stem cells (MSCs) remain poorly defined owing to their heterogeneity, the inability to assess in vivo self-renewal and the scarcity of markers allowing their identification, isolation and genetic manipulation. In the bone marrow (BM) of Nestin (Nes)-Gfp transgenic mice, CD31− CD45− GFP+ peri-vascular cells expressing endogenous nestin are associated with hematopoietic stem cells (HSCs) and innervated by fibers from the sympathetic nervous system (SNS). Flow cytometry sorting of BM CD45− Nes:GFP+ and CD45− Nes:GFP− cells has revealed that Nes:GFP+ cells, despite their rarity (4.0 ± 0.6% CD45− cells), contain all the colony-forming unit-fibroblastic (CFU-F) activity and have the exclusive capacity of forming self-renewing, multipotent clonal spheres that differentiate robustly along osteoblastic, chondrocytic and adipocytic lineages. To test in vivo self-renewal, single spheres derived from Nes-Gfp / Col2.3-Cre / R26R triple-transgenic animals were allowed to attach to phosphocalcic ceramic ossicles that were subcutaneously implanted into littermate mice that did not carry the transgenes. Histological analyses after 2 months revealed the presence of β-galactosidase+ osteoblasts (OBs) derived from Nes:GFP+ cells and not from 30,000 control CD45− Nes:GFP− cells. Hematopoietic areas were associated with Nes:GFP+ cells, that yielded per ossicle 310 ± 32 GFP+ secondary spheres (n=6), 38.6 ± 1.9% of which showed spontaneous multilineage differentiation into Col2.3+ OBs and Oil Red O+ adipocytes. Single secondary spheres subjected to a subsequent round of transplantation yielded after 8 months 8,557 ± 537 GFP+ spheres per ossicle (n = 7), which also generated Col2.3+ OBs, as a further proof of their self-renewal, osteoblastic differentiation potential and donor origin. Lineage-tracing studies in Nes-Cre / R26R mice have revealed the contribution of nestin-expressing cells in endochondral and membranous ossification. Administration of tamoxifen to adult Nes-CreERT2 mice bred to different reporter lines revealed that adult nestin-expressing BM cells could generate OBs, chondrocytes and osteocytes after 8-month chasing, suggesting an active role for adult nestin+ MSCs in physiological bone turnover. Genome-wide comparison analyses have shown that BM CD45− Nes:GFP+ cells are distinct from other stem cells but closest to in vitro expanded MSCs. Applying gene ontology analyses, metabolic and cell cycle genes were up- and down-regulated, respectively, in BM CD45− Nes:GFP+ cells. We have studied gene regulation, cell cycle and fate in response to granulocyte-colony stimulating factor (G-CSF), parathormone (PTH) and signals from the SNS, stimuli that regulate both hematopoietic and mesenchymal lineages in the BM. Cell cycle studies from FACS-sorted, flushed BM samples have confirmed that CD45− Nes:GFP+ cells are much more quiescent (90% G0/G1) than CD45− Nes:GFP− cells (58% G0/G1) but are selectively induced to proliferate after chemical sympathectomy (61% G0/G1) or PTH (70% G0/G1) administration in mice (n = 4–5). The inhibitory effects of the SNS and G-CSF (95% G0/G1) on BM CD45− Nes:GFP+ cells were not limited to cell cycle but also involved osteoblastic differentiation and expression of HSC maintenance genes. By contrast, in vivo or in vitro treatment with PTH selectively induced proliferation and osteoblastic differentiation of CD45− Nes:GFP+ cells, which express PTH receptor 1. We generated selective cell depletion models by intercrossing Nes-Cre and Nes-CreERT2 mice with a Cre-inducible diphtheria toxin receptor line (iDTR). In both models, HSC numbers decreased by ∼ 50% in the BM and increased in the spleen, an effect directly caused by selective BM cell depletion, as per in vitro experiments. In the more specific Nes-CreERT2 model, this effect was specific for HSCs and not for more mature progenitors. Cell depletion in Nes-Cre / iDTR and Nes-CreERT2 / iDTR mice reduced homing of hematopoietic progenitors by 73 and 90%, respectively. Finally, combined two-photon and confocal microscopy of the calvarial BM has demonstrated that highly purified, labeled HSCs rapidly (≤ 2h) home near Nes:GFP+ cells. Thus, cytokines, hormones, and the SNS regulate both HSC maintenance and bone formation in the BM stem cell niche through direct control of nestin-expressing MSCs. These results uncover an unprecedented partnership between two distinct somatic stem cell types and argue for a unique peri-vascular niche in the BM formed by MSC-HSC pairs. Disclosures: Scadden: Fate Therapeutics: Consultancy. Frenette:Glycomimetic: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document