scholarly journals Extracellular Vesicles Secreted by Human Urine-Derived Stem Cells Promote Ischemia Repair in a Mouse Model of Hind-Limb Ischemia

2018 ◽  
Vol 47 (3) ◽  
pp. 1181-1192 ◽  
Author(s):  
Qingwei Zhu ◽  
Qing Li ◽  
Xin Niu ◽  
Guowei Zhang ◽  
Xiaozheng Ling ◽  
...  

Background/Aims: Our previous studies have shown that human urine-derived stem cells (USCs) have great potential as a cell source for cytotherapy and tissue engineering and that extracellular vesicles (EVs) secreted by USCs (USCs-EVs) can prevent diabetes-induced kidney injury in an animal model. The present study was designed to evaluate the effects of USCs-EVs on ischemia repair. Methods: USCs-EVs were isolated and purified by a battery of centrifugation and filtration steps. The USCs-EVs were then characterized by transmission electron microscopy, western blot and tunable resistive pulse sensing techniques. After intramuscularly transplanting USCs-EVs into an ischemic mouse hind-limb, we observed the therapeutic effects of USCs-EVs on perfusion by laser doppler perfusion imaging, angiogenesis and muscle regeneration by histology and immunohistochemistry techniques over 21 days. We subsequently tested whether USCs-EVs can induce the proliferation of a human microvascular endothelial cell line HMEC-1 and a mouse myoblast cell line C2C12 by cell counting kit 8 assay in vitro. Meanwhile, the potential growth factors in the USCs-EVs and supernatants of the USCs cultures were detected by enzyme-linked immunosorbent assay. Results: The USCs-EVs were spherical vesicles with a diameter of 30–150 nm and expressed exosomal markers, such as CD9, CD63 and Tsg101. Ischemic limb perfusion and function were markedly increased in the hind-limb ischemia (HLI) model after USCs-EVs administration. Moreover, angiogenesis and muscle regeneration levels were significantly higher in the USCs-EVs treatment group than in the PBS group. The in vitro experiments showed that USCs-EVs facilitated HMEC-1 and C2C12 cell proliferation in a dose-dependent manner. Conclusions: These results revealed for the first time that USCs-EVs efficiently attenuate severe hind-limb ischemic injury and represent a novel therapy for HLI.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4233-4233
Author(s):  
Jeong-A Kim ◽  
Chang -Hoon Lee ◽  
Jin-A. Yoon ◽  
Woo-Sung Min ◽  
Chun-Choo Kim

Abstract We examined whether the injection of bone marrow mononuclear cells (BM-MNCs) or mesenchymal stem cells (MSCs) might augment angiogenesis and collateral vessel formation in a mouse model of hind limb ischemia. C57BL/6 BM-MNCs were isolated by centrifugation through a Histopaque density gradient and MSCs were obtained from C57BL/6 bone marrow and cultured in low-glucose DMEM media. Unilateral hind limb ischemia was surgically induced in C57BL/6 mice (control; n=4), and autologous BM-MNCs (Group 1; n=4, 1.8±0.2 x107/animal) or MSCs (Group 2; n=4, 1.0±0.14 x106/animal) or BM-MNCs and MSCs (Group 3; n=4, 2.3±0.1 x107 and 1.1±0.21 x106/animal) were transplanted into the ischemic tissue. Six weeks after transplantation, the group 1, group 2 and group 3 had a higher capillary/muscle ratio (0.82±0.12 vs 0.85±0.08 vs 0.97 ±0.03) than control (0.46±0.12, p<0.05) (Fig. 1). This result suggested that direct local transplantation of autologous BM-MNCs or MSCs seems to be a useful strategy for therapeutic neovascularization in ischemic tissues. Next, we evaluated whether bone marrow derived stem cells were participated in the process of local injected stem cells forming new vessels. In general, mobilizing stem cells from bone marrow to local site, MMP-9 has been known as an important molecule. So we used the MMP-9 deficient KO mice and wild type, 129SvEv mice were used in the experiments. Autologous BM-MNCs and MSCs were transplanted into the ischemic limb in MMP-9 (−/−) (n=4) after unilateral hind limb ischemia was surgically induced and then the same experiments was done in MMP-9 (+/+) mice (n=4). The number of the injected BM-MNCs and MSCs was 2.2±0.05 x107 and 0.87±0.17 x106/animal in MMP-9 (−/−). And the number of the injected BM-MNCs and MSCs was 2.1±0.17 x107 and 0.98±0.09 x106/animal in MMP-9 (+/+). No difference was seen in the BM-MNCs and MSCs were injected or not (0.52±0.07 vs 0.49±0.03,) in MMP-9 (−/−). But, in the case that BM-MNCs and MSCs were injected, the higher capillary/muscle ratio was seen in MMP-9 (+/+) compared to control (0.86 ±0.09 vs 0.49±0.03, P<0.05) (Fig 2). This data indicated that the mobilization of bone marrow derived stem cells would have an important role in the neovasculrization although the stem cells were injected directly into the muscle of ischemic limb. Figure Figure Figure Figure


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3697-3697
Author(s):  
Eun-Sun Yoo ◽  
KiHwan Kwon ◽  
Jee-Young Ahn ◽  
Soo-Ah Oh ◽  
Hye-Jung Chang ◽  
...  

Abstract Backgroud: Human umbilical cord blood (UCB) contains a high number of endothelial progenitor cells (EPCs) and may be useful for the treatment of ischemic disease. Recently, we have isolated EPCs from UCB having different biologic properties for angiogenic capabilities in vitro. In this present study, the aim is to examine the usefulness of OECs in hind-limb ischemia. Methods: Mononuclear cells from UCB cultured using EGM-2 medium with VEGF, IGF-1 and FGF for 21 days. Early spindle-shaped cells (early OECs), which were grown during the first week of culture and late cobblestone shaped cells (late OECs), which were in peak growth during the third week of culture were found. The hind-limb ischemia was established as follows: Athymic nude mice (BALB/C-nu) 18–22 g in weight were anesthetized with pentobarbital (60 mg/kg) and their left femoral arteries and main extension arteries were operatively resected. To examine the effect of the vasculogenesis of the two types of OECs, the mice were divided into three groups (PBS, early and late OECs). Twenty-four hours after operative excision 5 × 105 OECs in 200 μl and an equal volume of PBS were administered by intramuscular injection into the mice on hind-limb ischemia. To compare the effect of OECs on neovascularization in vivo, the analysis of blood flow of ischemic and healthy hind limbs was performed on days 1 and 21 after surgery using near-infrared (NIR) imaging with incocyanne Green (ICG). Results: Late OECs expressed a high level of mRNA on endothelial marker genes and formed capillary tubes in Matrigel plates. The early spindle cells excreted more angiogenic cytokines and had more migratory ability. We divided the mice into two groups according to the degree of perfusion; good (22.5–50%/min) and poor (0–22.5%) perfusion. OECs improved the blood flow of the ischemic hind-limb in the ’good’ perfusion group but not in the ’poor’ perfusion group. Early OECs led to a more significant improvement in blood flow than that of the late OECs. Conclusion: The different types of OECs from UCB have different biologic properties in vitro and different vasculogenic potential in vivo as well. The results might have potential application for the treatment of hind-limb ischemia.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 102-102
Author(s):  
Badr Kilani ◽  
Juliana Vieira Dias ◽  
Virginie Gourdou-Latyszenok ◽  
Eric Lippert ◽  
Raj Sewduth ◽  
...  

Abstract Background: Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders, characterized by significant increase in one or more myeloid lineages. A mutation in the Janus kinase 2 (JAK2) gene, JAK2V617F, was identified in half of the patients with Philadelphia chromosome-negative (Ph-) MPNs. This activating mutation causes hypersensitivity to certain growth factors, which explain the increased proliferation of myeloid progenitors. It has been reported that patients with MPNs have an increased risk of thrombosis but also increased microvessel density that may reflect angiogenesis in the spleen and bone marrow (Medinger, Br J Haematol, 2009), with no clear physiopathological explanation. Several recent studies have demonstrated the presence of JAK2V617F not only in blood cells but also in endothelial cells (EC) in these patients (Sozer, Blood, 2009; Teofili, Blood, 2011; Rosti, Blood, 2012). Hypothesis: We hypothesized that the presence of JAK2V617F in EC could change their properties leading to an increased angiogenesis process in MPNs. Methods: To determine whether the presence of JAK2V617F in EC was responsible for increased angiogenesis, we used an in vitro approach with human JAK2V617F ECs and an in vivo mouse model. We first used HUVECs (human umbilical vein endothelial cells) transduced with GFP lentivirus encoding human JAK2V617F. An empty lentivirus encoding only for GFP was used as a negative control. Proliferation of HUVECs was quantified during 3 days culture in EGM-2 medium and tube formation after culture in Matrigel™ was assessed by microscopy. Cell migration was determined by microscopy after scratch assay. Proteins expression level in cell lysate and supernatant was determined using Proteome ProfilerTM Array (R&D). The intensity of dot blots was determined by imageJ. For the in vivo approach, we crossed JAK2Flex/WT mice with PDGFβcreERT2 mice to generate endothelial-specific JAK2V617F knock-in mouse line (PDGFβERT2-JAK2 V617F/WT). Our lab was pioneer in developing microCT vascular imaging in order to precisely measure arterial vessel density and organization in 3 dimensions. To analyze neoangiogenesis (in the setting of ischemia), we used the model of mouse hind limb ischemia (Oses, ATVB, 2009): 11 days after ligature of the femoral artery, mice are perfused with latex labeled with barium and the hind limb vascular network is visualized with microCT. Results: We first observed that JAK2V617F HUVECs proliferate more than controls (coefficient rate of 3,53+/-0,18 versus 1,98 +/-0,05), reminding the phenotype of JAK2V617F hematopoietic cells. We then showed that JAK2V617F HUVECs are able to form more tubular structures in Matrigel™. Using an in vitro cell migration assay, we observed that JAK2V617F HUVECs invaded 45% (+/-2.3%) of the total surface area versus 27% (+/-3.3%) for controls. To confirm these results obtained in vitro, we analyzed the vascular network after hind limb ischemia in mice expressing JAK2V617F mutation specifically in endothelial cells (PDGFβERT2-JAK2V617F/WT). Our first results show an increased neoangiogenesis and further results will be presented at the conference. In order to decipher the mechanism responsible for the increased angiogenesis, we then quantified the expression of proteins that regulate angiogenesis, either in transduced HUVECs or in the culture supernatant. Interestingly, we have demonstrated a greater secretion of angiogenin, PDGF-AA, Endostatin, IGFBP-1, MCP-1 and CXCL-16 by HUVEC JAK2V617F. Discussion: In summary, our data suggest that the presence of JAK2V617F mutation in EC modifies their properties toward a pro-angiogenic profile and can explain, at least in part, the reported increase of angiogenesis in MPN patients. Once the role of JAK2V617F mutation in modifying EC properties will be confirmed, further work will be required to identify the molecular mechanisms underlying these phenotypic changes. It will also be particularly important to investigate if ECs are involved in the pathogenesis of increased angiogenesis observed in other diseases. Thus, they could be a new target in the treatment of pathological neo-angiogenesis. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 28 (20) ◽  
pp. 1384-1397 ◽  
Author(s):  
Zhecun Wang ◽  
Liang Zheng ◽  
Chong Lian ◽  
Yunling Qi ◽  
Wen Li ◽  
...  

Circulation ◽  
2008 ◽  
Vol 118 (suppl_18) ◽  
Author(s):  
Marie-Ange Renault ◽  
Jerome Roncalli ◽  
Joern Tongers ◽  
Hiromichi Hamada ◽  
Tina Thorne ◽  
...  

Gli transcription factors are mediators of hedgehog signaling and have been shown to be critical in several steps during development. In addition, Gli transcription factors have also been implicated as potent oncogenes. We have shown that the Hedgehog pathway is reactivated in the adult cardiovascular system under ischemic conditions and that the hedgehog pathway can be exploited therapeutically in ischemic disease; however the role and expression of the Gli transcription factors in the response to injury following ischemia have not been characterized. We induced hind-limb ischemia by resection of the left femoral artery of FVB mice and found that Gli2 and Gli3 mRNA were overexpressed in the ischemic tissue up to 4-fold and 9-fold respectively while Gli1 expression was not modulated. Immunostaining revealed that several cell types overexpressed Gli2 and Gli3 including myocytes and endothelial cells (ECs). To investigate the role and mechanism of action of Gli2 and Gli3, in vitro experiments were first performed. Overexpression of Gli2 and Gli3 promotes myoblast survival and proliferation. In ECs, overexpression of Gli2 and Gli3 promote survival and migration as well as induce expression of proteins involved in angiogenesis such as MMP-9 and osteopontin. To confirm the hypothesis of a favorable role of Gli2 and Gli3 in neovascularization and tissue repair following ischemic injury, adenovirus encoding LacZ, Gli2 and Gli3 were administrated locally in the tibialis anterior muscle, 2 days before hind limb ischemia surgery was performed. Both Ad-Gli2 and Ad-Gli3 treated mice exhibited higher proliferation in the regenerating muscle 7 days after ischemia. Moreover, Ad-Gli3 treated mice display higher capillary density (38.9±10.2 vessels/HPF vs. 26.1±6.1 in the control mice, p=0.012) and limb perfusion (the ratio of the perfusion of the ischemic leg vs. the control leg was 1.07±0.20 vs. 0.62±0,16 for the control mice, p=0.0003) 7 and 14 day after ischemia respectively. These data indicate that Gli2 and Gli3, transcription factors of the hedgehog pathway, are overexpressed in ischemic tissue and participate in tissue repair by promoting both neovascularization and proliferation and survival of myoblasts.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 217-217
Author(s):  
Ivana Rosova ◽  
Jan A. Nolta

Abstract Disorders such as peripheral artery disease cause hypoxic areas in tissues. Work from our group and others shows that stem cells appear to have innate mechanisms to respond to hypoxic conditions by migrating to the region of damage, and releasing trophic factors which initiate regeneration. Many tissues activate hepatocyte growth factor (HGF) as a response to ischemic injury. Multiple progenitor cell types express cMet, an HGF receptor. Mesenchymal stem cells (MSC) have been shown to improve regeneration of injured tissues in vivo, but their mechanisms of homing to the site of injury remain unclear. In the current studies we examined the potential for human MSC to repair injury caused by hind limb ischemia in immune deficient mice. We observed that hypoxic pre-conditioning of MSC upregulated expression of cMet, which could render the MSC more responsive to active HGF present at the site of ischemic injury. We first analyzed muscle lysates from mice that had undergone hind limb ischemia, vs sham-operated controls. ELISA results demonstrated that although a sham surgery caused a slight elevation in HGF levels 12 to 48 hours post surgery, ischemia caused a steady increase in HGF secretion from 12 hours to 48 hours post surgery. These data suggested that HGF might play a role in recruiting c-met+ MSC to the injury area. We next subjected primary human MSC to a 24-hr preconditioning in hypoxic (2 to 3% oxygen- actually tissue normoxia) vs. normoxic (21% oxygen, most commonly used in the incubator) conditions. MSC upregulated cMet in hypoxic conditions and then responded more robustly to HGF stimulation by signaling through cMet. Hypoxic pre-conditioning also caused signaling through a pro-survival Akt pathway, possibly improving the survival potential of MSC as they migrate in vivo. We next asked whether MSC are more motile in hypoxia. MSC were cultured in hypoxic or normoxic conditions +/− 25ng/ml HGF. While both HGF alone and a combination of hypoxia and HGF increased the cell migration capacity, treatment with hypoxia alone caused MSC to be the most migratory. These results suggest that hypoxic pre-conditioning may help MSC to migrate to the site of injury, while high active HGF levels in the tissue will hold the stem cells at the site of damage. Finally, to address the question of whether hypoxic pre-conditioning of MSC improves their tissue regeneration ability, we cultured them in hypoxic vs. normoxic conditions for 24 hrs and then transplanted them into NOD/SCID/B2m null mice that had undergone hind limb ischemia surgery one day prior to the transplant. Laser Doppler imaging showed significantly better blood flow recovery in the limbs of injured mice that were treated with pre-conditioned MSC, as compared to the saline control group. Mice that had received hypoxic pre-conditioned MSC improved bloodflow to the injured limb more rapidly than those transplanted with normoxic MSC, with a significant difference observed at day 5, demonstrating that hypoxic pre-conditioning increased the therapeutic potential of MSC. In summary, our data confirm that a 24 hour hypoxic pre-conditioning in vitro prior to transplantation improves the therapeutic potential of MSC, through activation of the pro-survival Akt pathway, upregulation of cMet, which allows them to be more responsive to the HGF activated at the site of ischemic injury, and an increased motility that allows them to more rapidly reach the area of injury.


Sign in / Sign up

Export Citation Format

Share Document