Electroactive nanoparticles loaded Silk protein/Chitosan macromolecular injectable hydrogel to improve therapeutic efficacy of mesenchymal stem cells in functional recovery after ischemic myocardial infarction

Author(s):  
Zheng Wu ◽  
Shujuan Cheng ◽  
Shaoping Wang ◽  
Wenzheng Li ◽  
Jinghua Liu

Abstract Background Currently, cardiac regeneration by stem cell-based tissue engineering is considered an important strategy for overcoming myocardial infarction. Therefore, this study is designed to explore the potential for differentiation of gold nanoparticles loaded injectable Silk protein/Chitosan hydrogel along with mesenchymal stem cells towards a cardiomyogenic phenotype. Methods The incorporated gold nanoparticles into chitosan-silk fibroin hydrogel (Au@Ch-SF) was validated by various analysis including FT-IR, NMR, XRD and SEM analysis. The major properties of Au@Ch-SF hydrogel such as weight loss, mechanical test and drug releasing activities also investigated. Further, the mesenchymal stem cells (MS) were encapsulated into hydrogel by incubating the MS cells with 100 µg/mL of Au@Ch-SF hydrogel in a humidified incubator at 37°C for 3 days in the presence of 5% CO2. In vitro toxicity effect of MS loaded Au@Ch-SF hydrogel was tested against cardiac myoblast H9C2 cells. Further, the tissue regenerative activities in myocardial infraction rats were examined by histology, apoptosis, and Cx43 cardiac-specific marker analysis. Results The gel formation time of Au@Ch-SF was comparatively lower than Ch and Ch-SF hydrogels which demonstrates the stronger intermolecular interactions between Ch and SF. The toxicity study showed that the prepared MS loaded Au@Ch-SF hydrogels did not possess toxicity against cardiac myoblast H9C2 cells. Further, the myocardial infarction rats were treated with MS loaded Au@Ch-SF hydrogel promotes the cardiac muscle fibers regeneration performance which was confirmed by β-MHC and Cx43 cardiac markers. Conclusions We demonstrate for the first time that encapsulation of MS with Au@Ch-SF hydrogels could promotes tissue regenerative activity in myocardial infraction tissues. The findings of this study suggest that MS encapsulated Au@Ch-SF hydrogels might be useful in the treatment of myocardial infarction.

2021 ◽  
Vol 12 (6) ◽  
Author(s):  
HuiYa Li ◽  
DanQing Hu ◽  
Guilin Chen ◽  
DeDong Zheng ◽  
ShuMei Li ◽  
...  

AbstractBoth weak survival ability of stem cells and hostile microenvironment are dual dilemma for cell therapy. Adropin, a bioactive substance, has been demonstrated to be cytoprotective. We therefore hypothesized that adropin may produce dual protective effects on the therapeutic potential of stem cells in myocardial infarction by employing an adropin-based dual treatment of promoting stem cell survival in vitro and modifying microenvironment in vivo. In the current study, adropin (25 ng/ml) in vitro reduced hydrogen peroxide-induced apoptosis in rat bone marrow mesenchymal stem cells (MSCs) and improved MSCs survival with increased phosphorylation of Akt and extracellular regulated protein kinases (ERK) l/2. Adropin-induced cytoprotection was blocked by the inhibitors of Akt and ERK1/2. The left main coronary artery of rats was ligated for 3 or 28 days to induce myocardial infarction. Bromodeoxyuridine (BrdU)-labeled MSCs, which were in vitro pretreated with adropin, were in vivo intramyocardially injected after ischemia, following an intravenous injection of 0.2 mg/kg adropin (dual treatment). Compared with MSCs transplantation alone, the dual treatment with adropin reported a higher level of interleukin-10, a lower level of tumor necrosis factor-α and interleukin-1β in plasma at day 3, and higher left ventricular ejection fraction and expression of paracrine factors at day 28, with less myocardial fibrosis and higher capillary density, and produced more surviving BrdU-positive cells at day 3 and 28. In conclusion, our data evidence that adropin-based dual treatment may enhance the therapeutic potential of MSCs to repair myocardium through paracrine mechanism via the pro-survival pathways.


2020 ◽  
Vol 2020 ◽  
pp. 1-14
Author(s):  
Jun Zhang ◽  
Ziming Liu ◽  
Yuwan Li ◽  
Qi You ◽  
Jibin Yang ◽  
...  

Background. FGF-2 (basic fibroblast growth factor) has a positive effect on the proliferation and differentiation of many kinds of MSCs. Therefore, it represents an ideal molecule to facilitate tendon-to-bone healing. Nonetheless, no studies have investigated the application of FGF-2-induced human amniotic mesenchymal stem cells (hAMSCs) to accelerate tendon-to-bone healing in vivo. Objective. The purpose of this study was to explore the effect of FGF-2 on chondrogenic differentiation of hAMSCs in vitro and the effect of FGF-2-induced hAMSCs combined with a human acellular amniotic membrane (HAAM) scaffold on tendon-to-bone healing in vivo. Methods. In vitro, hAMSCs were transfected with a lentivirus carrying the FGF-2 gene, and the potential for chondrogenic differentiation of hAMSCs induced by the FGF-2 gene was assessed using immunofluorescence and toluidine blue (TB) staining. HAAM scaffold was prepared, and hematoxylin and eosin (HE) staining and scanning electron microscopy (SEM) were used to observe the microstructure of the HAAM scaffold. hAMSCs transfected with and without FGF-2 were seeded on the HAAM scaffold at a density of 3×105 cells/well. Immunofluorescence staining of vimentin and phalloidin staining were used to confirm cell adherence and growth on the HAAM scaffold. In vivo, the rabbit extra-articular tendon-to-bone healing model was created using the right hind limb of 40 New Zealand White rabbits. Grafts mimicking tendon-to-bone interface (TBI) injury were created and subjected to treatment with the HAAM scaffold loaded with FGF-2-induced hAMSCs, HAAM scaffold loaded with hAMSCs only, HAAM scaffold, and no special treatment. Macroscopic observation, imageological analysis, histological assessment, and biomechanical analysis were conducted to evaluate tendon-to-bone healing after 3 months. Results. In vitro, cartilage-specific marker staining was positive for the FGF-2 overexpression group. The HAAM scaffold displayed a netted structure and mass extracellular matrix structure. hAMSCs or hAMSCs transfected with FGF-2 survived on the HAAM scaffold and grew well. In vivo, the group treated with HAAM scaffold loaded with FGF-2-induced hAMSCs had the narrowest bone tunnel after three months as compared with other groups. In addition, macroscopic and histological scores were higher for this group than for the other groups, along with the best mechanical strength. Conclusion. hAMSCs transfected with FGF-2 combined with the HAAM scaffold could accelerate tendon-to-bone healing in a rabbit extra-articular model.


2021 ◽  
Author(s):  
Lan Luo ◽  
Chen Yan ◽  
Naoki Fuchi ◽  
Yukinobu Kodama ◽  
Xu Zhang ◽  
...  

Abstract Background: Radiation-induced heart disease have been reported, but the mechanisms remain unclear. Mesenchymal stem cells (MSCs), also resident in heart are highly susceptible to radiation. We examined the hypothesis that altered secretion of exosomes from MSCs as the triggers of radiation-induced heart disease. Methods: By exposing human placental tissue-derived MSCs to 5 Gy γ-rays, we will then isolate exosomes from the culture medium 48h later and use to evaluate the quantity and quality changes of exosomes from MSCs after radiation exposures. The biological effects of exosomes from irradiated MSCs on HUVEC and H9c2 cells were also examined. Results: Although the amount and size distribution of exosomes did not differ between the non-irradiated and irradiated MSCs, miRNA sequences indicated many up- or down-regulated miRNAs in irradiated MSCs-exosomes. In vitro experiments using HUVEC and H9c2 cells showed that irradiated MSCs-exosomes significantly decreased cell proliferation, but increased cell apoptosis and DNA damage. Moreover, irradiated MSCs-exosomes impaired the tube formation of HUVEC cells and induced calcium overload of H9c2 cells. Conclusions: Exosomes released from irradiated MSCs shows an altered miRNA profiling and harmful effect to damage heart cells, which provides new insight on the mechanism of radiation-related heart disease risks.


2021 ◽  
Author(s):  
Jian Zhang ◽  
Yao Lu ◽  
Yangming Mao ◽  
Yue Yu ◽  
Tianyu Wu ◽  
...  

Abstract Background: Mesenchymal stem cells (MSCs) activated with IFN-γ elicit more powerful physical effects. Exosomes (Exos) secreted from MSCs have protective against myocardial injury. The aim of this study was to investigate whether Exsos derived from IFN-γ-pretreated MSCs exhibit more potent cardioprotective function and the underlying mechanisms. Methods: Exos were isolated from MSCs (Ctrl-Exo) and IFN-γ-primed MSCs (IFN-γ-Exo) and were then delivered to H9c2 cells or human umbilical vein endothelial cells (HUVECs) in vitro under oxygen and glucose deprivation (OGD) condition or in vivo in an infarcted rat heart. RNA sequencing was to identify the different expressed functional transcription factor (TF). Quantitative reverse transcription-PCR (qPCR) was to confirm the upregulated TF and miRNA in IFN-γ-primed MSCs. Dual-luciferase reporter gene assay were to analyze the transcriptional regulation of miRNAs by STAT1. The target of miR-21-5p (miR-21) was disclosed by luciferase reporter assays and qPCR. The function of BTG2 was verified in vitro under OGD condition.Result: IFN-γ-Exo accelerated migration, tube-like structure formation, and prevented H9c2 from OGD-induced apoptosis. Similarly, IFN-γ-Exo leaded to further reduction in fibrosis size, reduced cardiomyocyte apoptosis and improved cardiac function compared to Ctrl-Exo. miR-21 was significantly upregulated in both IFN-γ-primed MSCs and IFN-γ-Exo. STAT1 transcriptionally induced miR-21 expression. Up-regulated miR-21 can inhibit the expression of BTG2. BTG2 promoted H9c2 cells apoptosis and reversed the protective effect of miR-21 under OGD environment.Conclusion: IFN-γ-Exo have enhanced therapeutic efficacy against acute MI possibly through promoting angiogenesis and anti-apoptotic effect through increasing the level of miR-21, which directly targeted on BTG2.


2010 ◽  
Vol 22 (1) ◽  
pp. 346 ◽  
Author(s):  
S. A. Choi ◽  
J. H. Lee ◽  
K. J. Kim ◽  
E. Y. Kim ◽  
X. X. Li ◽  
...  

The dog is biologically comparable with humans with respect to stem cell kinetics, haematopoietic demand, and responsiveness to cytokines. The availability of canine mesenchymal stem cells allows for the establishment of the dog as a large animal model for testing the safety and efficacy of mesenchymal stem cells replacement therapy. Large animal models, such as the dog, are invaluable for working out the practicalities of a therapeutic regimen in a complex system and for verifying established mechanistic theories. Therefore, canine stem cells present the potential for unique and exciting biological opportunities. Recent observations also indicate that stem cells derived from second-trimester amniocentesis are pluripotent, capable of differentiating into multiple lineages, including representatives of all 3 embryonic germ layers. Compared with embryonic stem cells, amniotic fluid stem cells can be obtained without destroying embryos, thus avoiding much ethical controversy. The aim of the current study was to investigate adipogenic, osteogenic, and chondrogenic in vitro differentiation potential of canine amniotic fluid-derived mesenchymal stem cells by biological characterization. We successfully isolated and characterized canine amniotic fluid-derived mesenchymal stem cells (cAFS). Expression of stem cell-specific marker OCT3/4, SOX2, and NANOG was confirmed by RT-PCR. Flow cytometric analysis showed that cAFS were positive for CD44, CD29, and CD90 but negative for CD34. Immunocytochemical analysis also showed the expression of alkaline phosphatase, SOX2, SSEA-1, and SSEA-4. Following incubation with specific adipogenic, osteogenic, and chondrogenic agents, cAFS stained positive by Oil Red O and Alizarin Red S, respectively. In conclusion, according to the preview studies on other mammalians, cAFS is an appropriate source of pluripotent stem cells. Here, we demonstrated that cAFS has a high adipogenic, osteogenic, and chondrogenic differentiation potential in vitro. Therefore, amniotic fluid might be a suitable alternative source of stem cells. This study was financially supported by KOSEF (grant #R01-2008-000-21076-0), research fund of Chungnam National University, and the Korean MEST, through the BK21 program for creative research in animal biotechnology.


2016 ◽  
Vol 28 (2) ◽  
pp. 244 ◽  
Author(s):  
H. Malik ◽  
V. Sharma ◽  
S. Saini ◽  
S. Guha ◽  
D. Malakar

The present study was carried out for isolation and culture of adipose tissue-derived mesenchymal stem cells of goat (gADSC) and dogs (1 dog was suffering from hip dysplasia and another dog from paraplegia) and their characterisation with different markers. Adipose tissue of goat and dog were aseptically isolated and treated with collagenase for 2 h in a CO2 incubator. The enzymatic digested cells were filtered through a 41-µm filter and cells were resuspended in cell culture flask containing medium DMEM/F12, 10% fetal bovine serum, and 50 μg mL–1 gentamycin. In vitro-cultured ADSC were characterised by amplification of mesenchymal stem cell (MSC)-specific surface marker genes of CD44, CD29, and CD166 in PCR and by immunocytochemistry of MSC-specific marker of CD44. For in vitro chondrogenesis, ADSC at passage 3 were incubated in DMEM/F12 containing 100 nM dexamethasone, 1.25 μg mL–1 BSA, and 10 ng mL–1 BMP-4 ITS (insulin-transferrin-selenium) for 3 wk. Chondrogenic differentiation cells were confirmed by Safranin O staining and positive expression of chondrocyte-specific marker genes Aggrecan: primers F-TTGGACTTTGGCAGAATACC and R-CTTCCACCAATGTCGTATCC, and Collagen II: primers F-AACCCTGGAACTGACGGAAT and R-CTCACCCGTTTGACCTTTCG in PCR. Dog ADSC-derived chondrocytes were aseptically injected at 1 × 106 cells kg–1 of BW into dogs with hip dysplasia and paraplegia. Both dogs recovered well after 1 month of autologous transplantation and were able to move freely. Then, 10 dogs having massive wounds were injected with heterologous undifferentiated mesenchymal stem cells at 1 × 106 cells kg–1 of BW and all dogs were cured in an average of 20 days. Then, the paralyzed and fractured dogs were further treated with undifferentiated MSC at 1 × 106 cells kg–1 of BW and most of the dogs were cured properly. These findings may have implications for defining the physiological roles of ADSC in arthritis, some orthopaedic problems, joint regeneration, and neurological disorders and several new applications leading to novel therapeutic opportunities.


2012 ◽  
Vol 111 (suppl_1) ◽  
Author(s):  
Georges Makhoul ◽  
Yu-Ting Ma ◽  
Minh Duong ◽  
Ray C J Chiu ◽  
Renzo Cecere

Purpose: Human bone marrow mesenchymal stem cells (hbmMSCs) have been studied extensively for myocardial regenerative therapy. However, such cells require invasive procreation and suffer from donor age-related declining quality. Recently, a more abundant resource of young MSCs has been isolated from an otherwise discarded organ: the human placenta mesenchymal stem cells (hpMSCs). In this study, we wanted to examine the survival, differentiation, and functionality of xenogeneic hpMSCs when implanted into an induced myocardial rat infarction. Methods: To inspect their stemness,hpMSCs underwent an In Vitro cardiac cell differentiation in a DMEM medium containing 5 Azacytidine. Additionally, hpMSCs were tested in a myocardial infarction animal model. Female Lewis rats (40 animals) underwent left coronary artery ligation. Animals were divided into 4 groups. Group 1 was injected with hpMSCs in the peri-infarct region. Groups 2 and 3 received hbmMSCs and In Vitro differentiated hbmMSCs into cardiomyocytes respectively. Cell free medium was injected in group 4. Echocardiography was performed at baseline, day 4, weeks 3, 6, and 9 after ligation. Myocardial tissues were harvested and studied immunohistochemically for specific muscular and cardiac markers (Actin and Troponin I) on weeks 6 and 10. Results: In Vitro differentiation into cardiomyocyte lineage was achieved with the hpMSCs. HpMSCs were detected within rat myocardium by week 6 after their implantation. These cells stained positively for Actin and Troponin I. Preliminary echocardiographic data show cardiac functional increase in group 1. Whether hpMSCs can provide a superior effect than hbmMSCs or differentiated hbmMSCs is being investigated. Conclusions: In Vitro studies indicated that the hpMSCs can be differentiated into cardiac cells. When implanted into the rat infarcted myocardium, hpMSCs survived and were able to differentiate into cardiomyocytes. These cells appeared to increase cardiac function in an induced myocardial infarction rat model. With the advantages of easy availability and young age, hpMSCs could be more suitable for myocardial regenerative therapy.


2015 ◽  
Vol 2015 ◽  
pp. 1-14 ◽  
Author(s):  
Kai Kang ◽  
Ruilian Ma ◽  
Wenfeng Cai ◽  
Wei Huang ◽  
Christian Paul ◽  
...  

Background and Objective.Exosomes secreted from mesenchymal stem cells (MSC) have demonstrated cardioprotective effects. This study examined the role of exosomes derived from MSC overexpressing CXCR4 for recovery of cardiac functions after myocardial infarction (MI).Methods. In vitro, exosomes from MSC transduced with lentiviral CXCR4 (ExoCR4) encoding a silencing sequence or null vector were isolated and characterized by transmission electron microscopy and dynamic light scattering. Gene expression was then analyzed by qPCR and Western blotting. Cytoprotective effects on cardiomyocytes were evaluated and effects of exosomes on angiogenesis analyzed.In vivo, an exosome-pretreated MSC-sheet was implanted into a region of scarred myocardium in a rat MI model. Angiogenesis, infarct size, and cardiac functions were then analyzed.Results. In vitro, ExoCR4significantly upregulatedIGF-1αand pAkt levels and downregulated active caspase 3 levelin cardiomyocytes. ExoCR4also enhanced VEGF expression and vessel formation. However, effects of ExoCR4were abolished by an Akt inhibitor or CXCR4 knockdown.In vivo, ExoCR4treated MSC-sheet implantation promoted cardiac functional restoration by increasing angiogenesis, reducing infarct size, and improving cardiac remodeling.Conclusions.This study reveals a novel role of exosomes derived from MSCCR4and highlights a new mechanism of intercellular mediation of stem cells for MI treatment.


Sign in / Sign up

Export Citation Format

Share Document