Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
World J Clin Cases ; 9(30): 8953-8966, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34786379

ABSTRACT

Vascular injury is a frequent pathology in coronary artery disease. To repair the vasculature, scientists have found that endothelial progenitor cells (EPCs) have excellent properties associated with angiogenesis. Over time, research on EPCs has made encouraging progress regardless of pathology or clinical technology. This review focuses on the origins and cell markers of EPCs, and the connection between EPCs and coronary artery disease. In addition, we summarized various studies of EPC-capturing stents and EPC infusion therapy, and aim to learn from past technology to predict the future.

2.
Chin Med J (Engl) ; 131(1): 69-74, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29271383

ABSTRACT

BACKGROUND: Adipose-derived stromal vascular fraction (ADSVF) can be applied to repair tendon and ligament tears. ADSVF treatment has a better therapeutic potential than adipose stem cells alone in promoting the healing of connective tissue injury in rabbit models. Magnetic resonance imaging (MRI) and biomechanical testing were used in this study to evaluate the efficiency of SVF in the healing of tendon-bone interface of a rotator cuff injury after reattachment. METHODS: A total of 36 rabbits were studied between March and June 2016, 18 rabbits received the SVF-fibrin glue (SVF-FG) treatment and the other 18 formed the control group. ADSVF was isolated from each rabbit. A bilateral amputation of the supraspinatus tendon and parallel reconstruction was also performed on all the 36 rabbits. Then, a mixture of SVF and FG was injected into the tendon-bone interface of the SVF-FG group, whereas the control group only received FG. The animals were randomly sacrificed at 4, 8, and 12 weeks after surgery (n = 6 per group), respectively. The shoulders were prepared for MRI scanning and analysis of biomechanical properties. Analyses of variance were performed using SPSS 13.0. RESULTS: MRI scanning showed that the signal-to-noise quotient of the SVF-FG group was not significantly higher than that of the control group at either 4 (20.1 ± 3.6 vs. 18.2 ± 3.4, F = 1.570, P = 0.232) or 8 weeks (20.7 ± 3.3 vs. 18.0 ± 3.0, F = 2.162, P = 0.117) posttreatment, and only became significant after 12 weeks (27.5 ± 4.6 vs. 22.1 ± 1.9, F = 4.968, P = 0.009). Biomechanical properties such as the maximum load, maximum strength, and the stiffness for the SVF-FG group were significantly greater than that for the control group at 8 weeks' posttreatment (maximum load: 166.89 ± 11.62 N vs. 99.40 ± 5.70 N, P < 0.001; maximum strength: 8.22 ± 1.90 N/mm vs. 5.82 ±0.68 N/mm, P < 0.010; and the stiffness: 34.85± 3.00 Pa vs. 24.57± 5.72 Pa, P < 0.010). CONCLUSION: Local application of ADSVF might lead to better tendon-bone healing in rabbit models.


Subject(s)
Adipose Tissue , Rotator Cuff Injuries/diagnostic imaging , Rotator Cuff Injuries/therapy , Adipose Tissue/cytology , Animals , Biomechanical Phenomena , Fibrin Tissue Adhesive , Magnetic Resonance Imaging , Male , Rabbits , Rotator Cuff Injuries/physiopathology , Stem Cell Transplantation , Treatment Outcome
3.
PLoS One ; 9(10): e109711, 2014.
Article in English | MEDLINE | ID: mdl-25329797

ABSTRACT

Endothelial progenitor cells (EPCs) are the major source of cells that restore the endothelium during reendothelialization. This study was designed to investigate whether Schlafen 1 (Slfn1) has an effect on the proliferation and tube formation of EPCs in vivo. Slfn1 was expressed in rat EPCs. The overexpression of Slfn1 suppressed the proliferation and tube formation of EPCs; conversely, the knockdown of Slfn1 by shRNA promoted the proliferation and tube formation of EPCs. Furthermore, when Slfn1 was overexpressed, the EPCs were arrested in the G1 phase of the cell cycle. In contrast, when Slfn1 was knocked down, the EPCs progressed into the S phase of the cell cycle. Additionally, the overexpression of Slfn1 decreased the expression of Cyclin D1, whereas the knockdown of Slfn1 increased the expression of Cyclin D1; these findings suggest that Cyclin D1 is downstream of Slfn1 in Slfn1-mediated EPC proliferation. Taken together, these results indicate a key role for Slfn1 in the regulation of EPC biological behavior, which may provide a new target for the use of EPCs during reendothelialization.


Subject(s)
Cell Cycle Proteins/metabolism , Endothelial Progenitor Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Proliferation , Gene Silencing , Intracellular Space/metabolism , Male , Protein Transport , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley
4.
Stem Cells Dev ; 21(3): 487-96, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-21361857

ABSTRACT

Endothelial progenitor cells (EPCs) play an important role in accelerating endothelial repair after vascular injury. The proliferation and migration of EPCs is a critical first step in restoring endothelial. However, mechanisms for modulating EPC proliferation and migration are still being elucidated. Our previous study found that transient receptor potential canonical-1 (TRPC1) is involved in regulating store-operated Ca(2+) entry in EPCs through stromal interaction molecule 1. Therefore, in the present study, we sought to further investigate the regulation of proliferation and migration of EPCs by TRPC1. We found that the silencing of TRPC1 by 2 different RNA interference methods suppressed the proliferation and migration of EPCs. In addition, knockdown of TRPC1 significantly reduced of the amplitude of store-operated Ca(2+) entry and caused arrest of the EPC cell cycle in G1 phase. Analysis of the expression of 84 cell cycle genes by microarray showed that 9 genes were upregulated and 4 were downregulated by >2-fold in EPCs following TRPC1 silencing. The genes with expression changes were Ak1, Brca2, Camk2b, p21, Ddit3, Inha, Slfn1, Mdm2, Prm1, Bcl2, Mki67, Pmp22, and Ppp2r3a. Finally, we found that a Schlafen 1-blocking peptide partially reversed the abnormal cell cycle distribution and proliferation induced by TRPC1 knockdown, suggesting that Schlafen 1 is downstream of TRPC1 silencing in regulating EPC proliferation. In summary, these findings provide a new mechanism for modulating the biological properties of EPCs and suggest that TRPC1 may be a new target for inducing vascular repair by EPCs.


Subject(s)
Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Gene Silencing , Stem Cells/cytology , TRPC Cation Channels/metabolism , Animals , Biomarkers/metabolism , Calcium/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Endothelial Cells/metabolism , Flow Cytometry , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation , Gene Knockdown Techniques , Rats , Stem Cells/metabolism , TRPC Cation Channels/genetics , Wnt Signaling Pathway
5.
Mol Cell Biochem ; 363(1-2): 135-45, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22139302

ABSTRACT

The enhancement of re-endothelialisation is a critical therapeutic option for repairing injured blood vessels. Endothelial progenitor cells (EPCs) are the major source of cells that participate in endothelium repair and contribute to re-endothelialisation by reducing neointima formation after vascular injury. The over-expression of the inhibitor of differentiation or DNA binding 1 (Id1) significantly improved EPC proliferation. This study aimed to investigate the effects of Id1 on the phosphatidylinositol-3-kinase (PI3K)/Akt/nuclear factor kappa B (NFκB)/survivin signalling pathway and its significance in promoting EPC proliferation in vitro. Spleen-derived EPCs were cultured as previously described. Id1 was presented at low levels in EPCs, and was rapidly up-regulated by stimulation with vascular endothelial growth factor. We demonstrated that transient transfection of Id1 into EPCs activated the PI3K/Akt/NFκB/survivin signalling pathway and promoted EPC proliferation. The proliferation of EPCs was extensively inhibited by silencing of endogenous Id1, and knockdown of Id1 expression led to suppression of PI3K/Akt/NFκB/survivin signalling pathway in EPCs. In addition, blockade by the PI3K-specific inhibitor LY294002, Akt inhibitor, the NFκB inhibitor BAY 11-7082, the survivin inhibitor Curcumin, or the survivin inhibitor YM155 reduced the effects of Id1 transfection. These results suggest that the Id1/PI3K/Akt/NFκB/survivin signalling pathway plays a critical role in EPC proliferation. The Id1/PI3K/Akt/NFκB/survivin signalling pathway may represent a novel therapeutic target in the prevention of restenosis after vascular injury.


Subject(s)
Cell Proliferation , Endothelial Cells/enzymology , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Differentiation Protein 1/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stem Cells/enzymology , Active Transport, Cell Nucleus , Animals , Cell Nucleus/enzymology , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cysteine-Rich Protein 61/metabolism , Endothelial Cells/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , NF-kappa B/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spleen/cytology , Spleen/enzymology , Stem Cells/drug effects , Survivin , Transfection , Vascular Endothelial Growth Factor A/metabolism
6.
Zhonghua Xin Xue Guan Bing Za Zhi ; 39(7): 649-53, 2011 Jul.
Article in Chinese | MEDLINE | ID: mdl-22088247

ABSTRACT

OBJECTIVE: To investigate the effect of stromal interaction molecule 1 (STIM1) silencing on EPCs cell cycle. METHODS: Rat bone marrow derived endothelial progenitor cells (EPCs) were isolated and cultured in L-DMEM with 20% FBS. Ad-si/rSTIM1 and Ad-hSTIM1 were then transfected into EPCs and the expression of STIM1 mRNA was detected by RT-PCR. The cell cycle was determined using flow cytometry analysis and intracellular free Ca2+ was measured using LSCM. Co-immunoprecipitation was performed to examine the interaction between STIM1 and TRPC1. Protein levels of inositol 1, 4, 5-trisphosphate were analyzed with ELISA assay. RESULTS: Forty-eight hours after transfection, the expression of STIM1 mRNA was significantly downregulated (0.37 +/- 0.02 vs. 1.00 +/- 0.02, P < 0.05) and intracellular free Ca2+ level was significantly reduced (34.07 +/- 4.10 vs. 86.51 +/- 14.12, P < 0.05) in Ad-si/rSTIM1 group compared with control group. The cell cycle was arrested at G1 phase [(90.91 +/- 1.10)% vs. (77.10 +/- 0.56)%, P < 0.05] and the store-operated channel entry was strikingly inhibited in EPCs after treatment with Ad-si/rSTIM1. However, cotransfection of Ad-hSTIM1 with Ad-si/rSTIM1 significantly reversed these responses. Interestingly, co-immunoprecipitation study showed that STIM1 co-precipitated with TRPC1, and IP3 levels measured by ELISA were similar among three groups (P > 0.05). CONCLUSION: siRNA-mediated knockdown of STIM1 inhibited EPCs proliferation by reducing intracellular free Ca2+ through TRPC1-SOC signaling pathway.


Subject(s)
Endothelial Cells/cytology , Gene Silencing , Membrane Proteins/genetics , Neoplasm Proteins/genetics , RNA, Small Interfering , Stem Cells/cytology , Adenoviridae/genetics , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Genetic Vectors , Rats , Stromal Interaction Molecule 1 , Transfection , Transient Receptor Potential Channels/metabolism
8.
Basic Res Cardiol ; 105(6): 713-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20830586

ABSTRACT

Endothelial progenitor cells (EPCs) contribute to the process of reendothelialization and prevent neointimal formation after vascular injury. The present study was designed to investigate whether the cysteine-rich 61 (CYR61, CCN1), an important matricellular component of local vascular microenvironment, has effect on EPCs differentiation and reendothelialization in response to vascular injury in rat. Following balloon injury, CCN1 was rapidly induced and dynamically changed at vascular lesions. Overexpression of CCN1 by adenovirus (Ad-CCN1) accelerated reendothelialization and inhibited neointimal formation in the early phase (day 14) after vascular injury (p < 0.05), while no effect was shown on day 21. Ad-CCN1 treatment increased the adhering EPCs on the surface of injured vessels on day 7, and the ratio of GFP- and vWF-positive area to the total luminal length on day 14 was 2.3-fold higher in the Ad-CCN1-EPC-transplanted group than in controls. Consistent with these findings, CCN1-stimulated EPC differentiation in vitro and 20 genes were found differentially expressed during CCN1-induced EPC differentiation, including Id1, Vegf-b, Vegf-c, Kdr, Igf-1, Ereg, Tgf, Mdk, Ptn, Timp2, etc. Among them, negative transcriptional regulator Id1 was associated with CCN1 effect on EPC differentiation. Our data suggest that CCN1, from the microenvironment of injured vessels, enhances reendothelialization via a direct action on EPC differentiation, revealing a possible new mechanism underlying the process of vascular repair.


Subject(s)
Carotid Artery Injuries/metabolism , Cell Differentiation , Cell Proliferation , Cysteine-Rich Protein 61/metabolism , Endothelial Cells/metabolism , Stem Cells/metabolism , Animals , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cell Adhesion , Cell Differentiation/genetics , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Disease Models, Animal , Endothelial Cells/pathology , Endothelial Cells/transplantation , Gene Expression Regulation , Inhibitor of Differentiation Protein 1/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation , Stem Cells/pathology , Time Factors , Transduction, Genetic
9.
Biochem Biophys Res Commun ; 398(2): 315-20, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20599714

ABSTRACT

Knockdown of stromal interaction molecule 1 (STIM1) significantly suppresses neointima hyperplasia after vascular injury. Endothelial progenitor cells (EPCs) are the major source of cells that respond to endothelium repair and contribute to re-endothelialization by reducing neointima formation after vascular injury. We hypothesized that the effect of STIM1 on neointima hyperplasia inhibition is mediated through its effect on the biological properties of EPCs. In this study, we investigated the effects of STIM1 on the proliferation and migration of EPCs and examined the effect of STIM1 knockdown using cultured rat bone marrow-derived EPCs. STIM1 was expressed in EPCs, and knockdown of STIM1 by adenoviral delivery of small interfering RNA (siRNA) significantly suppressed the proliferation and migration of EPCs. Furthermore, STIM1 knockdown decreased store-operated channel entry 48h after transfection. Replenishment with recombinant human STIM1 reversed the effects of STIM1 knockdown. Our data suggest that the store-operated transient receptor potential canonical 1 channel is involved in regulating the biological properties of EPCs through STIM1. STIM1 is a potent regulator of cell proliferation and migration in rat EPCs and may play an important role in the biological properties of EPCs.


Subject(s)
Cell Movement , Cell Proliferation , Endothelial Cells/physiology , Membrane Glycoproteins/physiology , Stem Cells/physiology , Animals , Gene Knockdown Techniques , Humans , Membrane Glycoproteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Rats , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL