Objective To develop a new method for a tissue engineered vascular graft by combining endothelial cells and an acelluarized allogenic matrix. Methods Acellularized matrix tubes were obtained by a 0.1% trypsin and 0 02% EDTA solution for 24 hours and 1% Triton X 100 for 176 hours, respectively. Endothelial cells were isolated from alloaorta and expanded in vitro. Finally, the inner surface of acellularized matrix was reseeded with endothelial cells. Acellularity and reseeding were analysed by light microscopy and scanning electron microscopy. Results The acellularization procedure resulted in an almost complete removal of the original cells and the loose three-dimensional (3D) matrix. The acellular matrix could be reseeded with expanded endothelial cells in vitro, and endothelial cells had the potential of spread and proliferation. Conclusion Acellular matrix produces by Tritoon X-100 and trypsin possesses satisfactory biocompatibility for allogenic endothelial cell. Vascular grafts can be generated in vitro by a combination of endothelial cells and allogenic acelluarized matrix.
Objective To observe the proliferation and migration of endothelial cells after 30% total burn surface area (TBSA) of deep partial thickness scald, and the effect of basic fibroblast growth factor (bFGF) on angiogenesis during wound healing.Methods A total of 133 male Wistar ratswere divided randomly into normal control (n=7), injured control group (n=42), bFGF group (n=42) andanti-c-fos group (n=42). The apoptosis expression of fibroblasts was determinedwith in situ hybridization and the changes of proliferation cell nuclear antigen(PCNA), focal adhesion rinase(FAK), c-fos and extracellular signalregulated kinase(ERK) proteins expression were detected with immunohistochemistry staining technique after 3 hours, 6 hours, 1 day, 3 days, 7 days, 14 days and 21 days of scald.Results In injured control group and bFGF group, theproliferation rate of the vascular endothelial had evident changes 7 days and14 days after scald; the expression of FAK was increased 14 days after scald. ERK proteins expression was different between injury control group and bFGF group at initial stage after scald. Stimulation of ERKs by bFGF led to up-regulation of c-fos and b expression of FAK. Conclusion Exogenous bFGF extended the influence on wound healing process by ERK signaling pathway, affecting migration cascade of vascular endothelial cell. The oncogene proteins play an important role on accelerating angiogenesis duringwound healing.
Abstract In order to investigate the mechanism ofregeneration of lymphatic vessel, the regulatory control of various cell factors on the new born bovine lymphatic endothelial cell (NBLEC) was observed. The cell factors used for investigation were bFGF, TGFα, EGF, TNFα and IL-1α. The results showed that bFGF, TGFα and EGF could stimulate NBLEC proliferation and DNA synthesis in dosage-dependent pattern. Combined use of either two factorsdid not increase the effect, and bFGF could increase cell migration and improve the activity of tissue plasminogen activator (t-PA). TNFα and IL-α inhibited NBLEC regeneration and DNA synthesis but TNFα improved the activity of t-PA. It could be concluded that growth factor and inflammatory factor had differentrole on regeneration of NBLEC, such as cell proliferation, migration and t-PA activity. bFGF was the main factor which improved the regenerationof lymphatic endothelial cell.
Objective To examine the effects of hypoxia on endothelial-to-mesenchymal transition of porcine pulmonary arterial endothelial cells ( PAECs) .Methods The porcine PAECs were divided into a normoxia group and a hypoxia group. The cells in two groups were exposed to normoxic or hypoxic condition for 1,4, and 7 days respectively. The immunofluorescence,Western blot and RT-PCR were used to detect the protein and mRNA expressions of VE-cadherin and α-SMA. Results The porcine primary PAECs formed typical monolayer of cobblestone appearance on normoxia condition, and had a spindle-shaped appearance on hypoxia condition. Immunofluorescence results showed that these PAECs expressed mesenchymal cells specific marker of α-SMA. With the hypoxic time prolongation, the ratio of transdifferentiated smooth musclelike cells from PAECs was gradually increased ( P lt; 0. 01) . Western blot assay demonstrated that the expression level of VE-cadherin protein and mRNA was reduced gradually, but the expression level of α-SMA protein and mRNA was increased. Conclusion Hypoxia can induce endothelial-to-mesenchymal transition, which may be involved in the development of a variety of diseases.
After escaping from the hyperacute rejection (HAR), the xenograft has to be faced the challenge of acute vascular, acute cellular and even chronic rejection. Endothelial cells have been confirmed as a kind of antigen processing cell (APC) in allo-rejection. The porcine aortic endothelial cell (PAEC) expressed SLA-II and B7 which are the characteristics of professional APC. PAEC also has plenty of alpha-Gal residues, whether the antigen play any role in the post-HAR is still unknown. Human and porcine peripheral blood lymphocyte (PBLC) were isolated and divided into two parts, one for the effectors and the another were incubated with mitomycin C (MMC) as stimulators. The two kinds of PBLC were mixed-cultured within five days. Cultured PAEC from NJZ Pig was incubated with MMC and divided into two: One digested with alpha-galactosidase. The two kinds of PAEC were taken as stimulators to mixed-culture with human PBLC for five days. All the proliferation was detected with 3H-TdR intermingled in the system. The results showed that allo-MLR was ber than xeno-MLR in the cases. The proliferation was much ber when PAEC was used as the stimulator than that of porcine PBLC. However, the response was remarkably decreased after the digestion of alpha-Gal with alpha-galactosidase. The conclusion was that the low response of porcine-to-human MLR in vitro might be related to the predominant indirect pathway of antigen recognition in this system. While PAEC was used as the stimulator the proliferation in MLR was ber which might be concerned that PAEC itself was an APC as well as xeno-antigen sources, thus the direct pathway was predominant and worked more efficiently. The alpha-Gal might induce T cell proliferation through the linkage with the biological big molecules working as a complete antigen. The other post-HAR antigen might also exist in PAEC such as SLA-II, etc.
The aim of the study is to identify the effects and underlying mechanisms of visfatin on inflammation and necroptosis in vascular endothelial cells. Human umbilical vein endothelial cells (HUVECs) were stimulated with visfatin or pretreated with Polyinosinic acid (LOX-1 inhibitor). By using the Western blot, RT-PCR, immunocytochemistry, enzyme-linked immunosorbent assay (ELISA), MTT and flow cytometry technique, the occurrence of inflammation and necroptosis in HUVECs were evaluated. Our results showed that 100 ng/mL visfatin significantly increased the mRNA and protein expression of monocyte chemotactic protein 1 (MCP-1) and LOX-1 after 24 hours’ treatment in HUVECs. However, pretreatment with Polyinosinic acid could significantly reduce the expression of MCP-1 compared with visfatin group. Additionally, 100 ng/mL visfatin could induce the production of necrotic features and increase the mRNA expression of BMF (one of the markers of necroptosis), while pretreating with Polyinosinic acid markedly downregulated the mRNA expression of BMF gene and promoted the cell proliferation. These results indicate that visfatin might induce inflammation and necroptosis via LOX-1 in HUVECs, suggesting that visfatin plays a central role in the development of atherosclerosis.
ObjectiveTo explore the effect of Kaempferol on bone microvascular endothelial cells (BMECs) in glucocorticoid induced osteonecrosis of the femoral head (GIONFH) in vitro. MethodsBMECs were isolated from cancellous bone of femoral head or femoral neck donated voluntarily by patients with femoral neck fracture. BMECs were identified by von Willebrand factor and CD31 immunofluorescence staining and tube formation assay. The cell counting kit 8 (CCK-8) assay was used to screen the optimal concentration and the time point of dexamethasone (Dex) to inhibit the cell activity and the optimal concentration of Kaempferol to improve the inhibition of Dex. Then the BMECs were divided into 4 groups, namely, the cell group (group A), the cells treated with optimal concentration of Dex group (group B), the cells treated with optimal concentration of Dex+1 μmol/L Kaempferol group (group C), and the cells treated with optimal concentration of Dex+5 μmol/L Kaempferol group (group D). EdU assay, in vitro tube formation assay, TUNEL staining assay, Annexin Ⅴ/propidium iodide (PI) staining assay, Transwell migration assay, scratch healing assay, and Western blot assay were used to detect the effect of Kaempferol on the proliferation, tube formation, apoptosis, migration, and protein expression of BMECs treated with Dex. ResultsThe cultured cells were identified as BMECs. CCK-8 assay showed that the optimal concentration and the time point of Dex to inhibit cell activity was 300 μmol/L for 24 hours, and the optimal concentration of Kaempferol to improve the inhibitory activity of Dex was 1 μmol/L. EdU and tube formation assays showed that the cell proliferation rate, tube length, and number of branch points were significantly lower in groups B-D than in group A, and in groups B and D than in group C (P<0.05). TUNEL and Annexin V/PI staining assays showed that the rates of TUNEL positive cells and apoptotic cells were significantly higher in groups B-D than in group A, and in groups B and D than in group C (P<0.05). Scratch healing assay and Transwell migration assay showed that the scratch healing rate and the number of migration cells were significantly lower in groups B-D than in group A, and in groups B and D than in group C (P<0.05). Western blot assay demonstrated that the relative expressions of Cleaved Caspase-3 and Bax proteins were significantly higher in groups B-D than in group A, and in groups B and D than in group C (P<0.05); the relative expressions of matrix metalloproteinase 2, Cyclin D1, Cyclin E1, VEGFA, and Bcl2 proteins were significantly lower in groups B-D than in group A, and in groups B and D than in group C (P<0.05). Conclusion Kaempferol can alleviate the damage and dysfunction of BMECs in GIONFH.
Objective To investigate the effect and potential mechanism of bone marrow mesenchymal stem cells (BMSCs) - derived extracellular vesicles (EVs) on lung tissue injury in mice with severe acute pancreatitis (SAP). Methods A total of 24 specific pathogen free grade male C57BL/6 mice and primary mouse lung microvascular endothelial cells (PMVECs) were selected. The mice were divided into sham group, SAP group, and BMSC group, with 8 mice in each group. The mouse primary PMVECs were divided into model group [sodium taurocholate (NaTC) group], BMSC-EV group, and control group. Extraction and characterization of healthy mouse BMSCs and their derived extracellular vesicles (BMSC-EVs) were conducted. A mouse model of SAP was established, and BMSC-EVs were injected into SAP mice by tail vein or intervened in PMVECs in vitro, to observe the pathological damage of pancreatic and lung tissues, the changes of serum amylase, lipase, and inflammatory factors [tumor necrosis factor α (TNF-α), interleukin-6 (IL-6)], the expression of inflammatory factors of lung tissues and PMVECs, and the endothelial cell barrier related proteins [E-cadherin, ZO-1, intercellular cell adhesion molecule-1 (ICAM-1)], and tight junctions between PMVECs to explore the effects of BMSC-EVs on pancreatic and lung tissues in SAP mice and PMVECs in vitro. Results BMSCs had the potential for osteogenic, chondrogenic, and lipogenic differentiation, and the EVs derived from them had a typical cup-shaped structure with a diameter of 60-100 nm. BMSC-EVs expressed the extracellular vesicle-positive proteins TSG101 and CD63 and did not express the negative protein Calnexin. Compared with the mice in the sham group, the SAP mice underwent significant pathological damage to the pancreas (P<0.05), and their serum amylase, lipase, inflammatory factor IL-6, and TNF-α levels were significantly up-regulated (P<0.05); whereas, BMSC-EVs markedly ameliorated the pancreatic tissue damage in the SAP mice (P<0.05), down-regulated the levels of peripheral serum amylase, lipase, IL-6 and TNF-α (P<0.05), and up-regulated the level of anti-inflammatory factor IL-10 (P<0.05). In addition to this, the SAP mice showed significant lung histopathological damage (P<0.05), higher neutrophils and macrophages infiltration (P<0.05), higher levels of the inflammatory factors TGF-β and IL-6 (P<0.05), as well as reduced barrier protein E-cadherin, ZO-1 expression and elevated expression of ICAM-1 (P<0.05). BMSC-EVs significantly ameliorated lung histopathological injury, inflammatory cells infiltration, inflammatory factor levels, and expression of barrier proteins, and suppressed ICAM-1 expression (P<0.05). In the in vitro PMVECs experiments, it was found that intercellular tight junctions were broken in the NaTC group, and the levels of inflammatory factors TNF-α and IL-6 were significantly up-regulated (P<0.05), the protein expression of E-cadherin and ZO-1 was significantly down-regulated (P<0.05), and the expression of ICAM-1 was significantly up-regulated (P<0.05). BMSC-EVs significantly improved intercellular tight junctions in the NaTC group and inhibited the secretion of TNF-α and IL-6 (P<0.05), up-regulated the expression of the barrier proteins E-cadherin and ZO-1, and down-regulated the expression of ICAM-1 (P<0.05). Conclusion BMSC-derived EVs ameliorate lung tissue injury in SAP mice by restoring the lung endothelial cell barrier and inhibiting inflammatory cell infiltration.
Coronary atherosclerotic heart disease is a serious threat to human life and health. In recent years, the main treatment for it is to implant the intravascular stent into the lesion to support blood vessels and reconstruct blood supply. However, a large number of experimental results showed that mechanical injury and anti-proliferative drugs caused great damage after stent implantation, and increased in-stent restenosis and late thrombosis risk. Thus, maintaining the integrity and normal function of the endothelium can significantly reduce the rate of thrombosis and restenosis. Stem cell mobilization, homing, differentiation and proliferation are the main mechanisms of endothelial repair after vascular stent implantation. Vascular factor and mechanical microenvironmental changes in implanted sites have a certain effect on re-endothelialization. In this paper, the process of injury caused by stent implantation, the repair mechanism after injury and its influencing factors are expounded in detail. And repairing strategies are analyzed and summarized. This review provides a reference for overcoming the in-stent restenosis, endothelialization delay and late thrombosis during the interventional treatment, as well as for designing drug-eluting and biodegradation stents.
Objective To explore the facilitative effects of different allogenic cells injected into the denervated muscles on the nerve regeneration, the protection of the myoceptor degeneration, and the promotion for rehabilitation of the muscular function. Methods Schwann cells, myoblast cells, and renal endothelial cells were prepared from 400 SD rats aged 7 days and weighing 20.0±2.3 g. Thirty-six adult female SD rats weighing 120-150 g were randomly divided into 4 groups(n=9). Under the asepsis condition, the left ischiadic nerves of all the SD rats were cut off, and the primary suture of the epineurium was performed. After operation, the different corresponding cells were injected into the triceps muscles of the rat calf in each group once per week for 4 times in all. One ml of Schwann cells (1×106/ml) was injected into the rats in Group A; 1 ml of the mixed cells of Schwann cells and myoblast cells (1×106/ml) was injected into the rats in Group B; 1 ml of the extract from the mixed cells of Schwann cells, myoblast cells, and renal endothelial cells (1×106/ml) was injected into the rats in Group C; 1 ml of the culture medium without any serum was injected into the rats in Group D as a control. After operation, observation was made for the general condition of the rats; 3 months after operation, enzymohistochemistry and the CJun expression were performedin the ventricornual motor neuron. At the proximal and the distal ends of the nerve suture, the density of neurilemma cells in the unit area and the area size of the regenerated nerve fibers were observed and measured. Results The affected limbs of the rats in Groups A, B and C improved 13 months after operation. The ulcers and swelling at the ankles gradually relieved and the rats could move normally 3 months after operation. However, the affected limbsof the rats in Group D still had ulcers and swelling, with an obvious contracture of the toes and a difficult movement. Three months after operation, the number of the target muscle myoceptor, the number of the Actin positive cells, the activity of the various enzymes in the denervated muscles, and the histological changes of the regenerated nerves were better in Group C than in Groups A and B (P<0.01); and they were all better in Groups A, B and C than in Group D(Plt;0.01). Conclusion Schwann cells, the mixture of Schwann cells and myoblast cells, and the extract from the mixture of Schwann cells, myoblast cells and renal endothelial cells can all promote neurotization and rehabilitation of the muscular function, and protect against the myoceptor degeneration. However, the effect of the extract is superior to that of Schwann cells or the mixed cells.