The expression of GFP was gradually decreased with prolonged treatment time. (A) Activities of fluorescent cells in MSC-ADM scaffolds transplanted into the wound site on days 3, 5, 7 and 14. Green and red represent the GFP +-MSCs and collagen in left and middle columns, respectively, and their merge in the right column. The wounds of control group imaged by TPLSCM showed the changes of collagen intensity on days 3, 5, 7 and 14. (Original magnification, 40x. Scale bar, 20 µm). (B) SHG intensity of collagen at different time points with increased treatment times in MSCADM group and control group. (C) Fluorescence intensity with treatment time. (D) Comparison of GFP levels in each group (3, 5, 7 and 14 days) via western blot analysis. (E) Statistical analysis of the expression of GFP. The data are expressed as the mean values, and the error bars represent SD (n = 5). *P<0.05, **P<0.01, ***P<0.001. 

The expression of GFP was gradually decreased with prolonged treatment time. (A) Activities of fluorescent cells in MSC-ADM scaffolds transplanted into the wound site on days 3, 5, 7 and 14. Green and red represent the GFP +-MSCs and collagen in left and middle columns, respectively, and their merge in the right column. The wounds of control group imaged by TPLSCM showed the changes of collagen intensity on days 3, 5, 7 and 14. (Original magnification, 40x. Scale bar, 20 µm). (B) SHG intensity of collagen at different time points with increased treatment times in MSCADM group and control group. (C) Fluorescence intensity with treatment time. (D) Comparison of GFP levels in each group (3, 5, 7 and 14 days) via western blot analysis. (E) Statistical analysis of the expression of GFP. The data are expressed as the mean values, and the error bars represent SD (n = 5). *P<0.05, **P<0.01, ***P<0.001. 

Source publication
Article
Full-text available
Background/aims: Bone marrow Mesenchymal stem cells (MSCs) are promising for promoting cutaneous wound healing through reinforcing cellular processes. We evaluated the effect of GFP-tagged MSCs transplantation on skin regeneration in excisional wounds in mice. Methods: MSCs from GFP-labeled transgenic mice were co-cultured with acellular dermal...

Contexts in source publication

Context 1
... observe the behavior of MSCs during wound repair, regenerative skin tissues located at the wound sites were collected on days 3, 5, 7 and 14 after MSC-ADM scaffolds were implanted into the skin wounds and immediately imaged by TPLSCM. In addition, the wounds of control group were also imaged by TPLSCM (Fig. 5A). By evaluating the GFP and SHG signal intensities of the corresponding samples from the photographs shown in Fig. 5A, we obtained two curves to depict the changes of the number of MSCs and collagen accumulation at different time points (Fig. 5C and Fig. 5B). The result revealed that the collagen content markedly increased in both two groups as treatment time was prolonged and the content of MSC-ADM group was higher than that of control group at the same healing day (Fig. 5B). The number of GFP+-MSCs gradually decreased (Fig. 5C) with time. This result was further confirmed by western blot analyses (Fig. 5D and 5E). The expression level of GFP was significantly decreased in the wounds and approached zero on days 14 after wound ...
Context 2
... observe the behavior of MSCs during wound repair, regenerative skin tissues located at the wound sites were collected on days 3, 5, 7 and 14 after MSC-ADM scaffolds were implanted into the skin wounds and immediately imaged by TPLSCM. In addition, the wounds of control group were also imaged by TPLSCM (Fig. 5A). By evaluating the GFP and SHG signal intensities of the corresponding samples from the photographs shown in Fig. 5A, we obtained two curves to depict the changes of the number of MSCs and collagen accumulation at different time points (Fig. 5C and Fig. 5B). The result revealed that the collagen content markedly increased in both two groups as treatment time was prolonged and the content of MSC-ADM group was higher than that of control group at the same healing day (Fig. 5B). The number of GFP+-MSCs gradually decreased (Fig. 5C) with time. This result was further confirmed by western blot analyses (Fig. 5D and 5E). The expression level of GFP was significantly decreased in the wounds and approached zero on days 14 after wound ...
Context 3
... observe the behavior of MSCs during wound repair, regenerative skin tissues located at the wound sites were collected on days 3, 5, 7 and 14 after MSC-ADM scaffolds were implanted into the skin wounds and immediately imaged by TPLSCM. In addition, the wounds of control group were also imaged by TPLSCM (Fig. 5A). By evaluating the GFP and SHG signal intensities of the corresponding samples from the photographs shown in Fig. 5A, we obtained two curves to depict the changes of the number of MSCs and collagen accumulation at different time points (Fig. 5C and Fig. 5B). The result revealed that the collagen content markedly increased in both two groups as treatment time was prolonged and the content of MSC-ADM group was higher than that of control group at the same healing day (Fig. 5B). The number of GFP+-MSCs gradually decreased (Fig. 5C) with time. This result was further confirmed by western blot analyses (Fig. 5D and 5E). The expression level of GFP was significantly decreased in the wounds and approached zero on days 14 after wound ...
Context 4
... observe the behavior of MSCs during wound repair, regenerative skin tissues located at the wound sites were collected on days 3, 5, 7 and 14 after MSC-ADM scaffolds were implanted into the skin wounds and immediately imaged by TPLSCM. In addition, the wounds of control group were also imaged by TPLSCM (Fig. 5A). By evaluating the GFP and SHG signal intensities of the corresponding samples from the photographs shown in Fig. 5A, we obtained two curves to depict the changes of the number of MSCs and collagen accumulation at different time points (Fig. 5C and Fig. 5B). The result revealed that the collagen content markedly increased in both two groups as treatment time was prolonged and the content of MSC-ADM group was higher than that of control group at the same healing day (Fig. 5B). The number of GFP+-MSCs gradually decreased (Fig. 5C) with time. This result was further confirmed by western blot analyses (Fig. 5D and 5E). The expression level of GFP was significantly decreased in the wounds and approached zero on days 14 after wound ...
Context 5
... observe the behavior of MSCs during wound repair, regenerative skin tissues located at the wound sites were collected on days 3, 5, 7 and 14 after MSC-ADM scaffolds were implanted into the skin wounds and immediately imaged by TPLSCM. In addition, the wounds of control group were also imaged by TPLSCM (Fig. 5A). By evaluating the GFP and SHG signal intensities of the corresponding samples from the photographs shown in Fig. 5A, we obtained two curves to depict the changes of the number of MSCs and collagen accumulation at different time points (Fig. 5C and Fig. 5B). The result revealed that the collagen content markedly increased in both two groups as treatment time was prolonged and the content of MSC-ADM group was higher than that of control group at the same healing day (Fig. 5B). The number of GFP+-MSCs gradually decreased (Fig. 5C) with time. This result was further confirmed by western blot analyses (Fig. 5D and 5E). The expression level of GFP was significantly decreased in the wounds and approached zero on days 14 after wound ...
Context 6
... observe the behavior of MSCs during wound repair, regenerative skin tissues located at the wound sites were collected on days 3, 5, 7 and 14 after MSC-ADM scaffolds were implanted into the skin wounds and immediately imaged by TPLSCM. In addition, the wounds of control group were also imaged by TPLSCM (Fig. 5A). By evaluating the GFP and SHG signal intensities of the corresponding samples from the photographs shown in Fig. 5A, we obtained two curves to depict the changes of the number of MSCs and collagen accumulation at different time points (Fig. 5C and Fig. 5B). The result revealed that the collagen content markedly increased in both two groups as treatment time was prolonged and the content of MSC-ADM group was higher than that of control group at the same healing day (Fig. 5B). The number of GFP+-MSCs gradually decreased (Fig. 5C) with time. This result was further confirmed by western blot analyses (Fig. 5D and 5E). The expression level of GFP was significantly decreased in the wounds and approached zero on days 14 after wound ...
Context 7
... a two-photon laser scanning confocal microscopy platform, we first detected GFP expression in the MSCs at different healing stages (3,5,7,14,21, and 28 days after transplantation). The results showed that few exogenous GFP + -MSCs were detected in the wound sites, and the numbers were not significantly different from our result using transfected MSCs (data not shown). To confirm this, quantitative measurement techniques were used to analyze the GFP expression in different samples. PCR and sequencing of the GFP gene revealed there were almost no exogenous MSCs or differentiated cells derived from the GFP+-MSCs that survived in the regenerative tissues after 14 days (Fig. 6). Western blotting confirmed this result (Fig. 5). Therefore, we focused on the time frame of 1 day to 2 weeks to investigate the functional mechanism of the exogenous MSCs in tissue regeneration. Thus, the wound healing process based on MSC therapy would be analogous to the natural healing process after 2 ...

Citations

... Based on the study, BM-MSCs migrated into skin wounds 9,10 and released bioactive components that attracted additional repair cells from the host animals. 11,12 Wu et al. 13 also have revealed that MSCs could differentiate into keratinocytes at the wound bed. Since bone marrow-derived cells (BM-MSCs) have been the subject of the majority of the research into MSC biology to date, BM-MSCs are usually recognized as the primary cell source for potential clinical applications. ...
Article
Full-text available
Background To maintain and enhance the wound healing effects of mesenchymal stem cells (MSCs), a scaffold for hosting MSCs is needed, which ought to be completely biocompatible, durable, producible, and of human source. Objective To build a cell-extracellular matrix (ECM) complex assembled by human umbilical cord mesenchymal stem cells (HuMSCs) and to investigate its clinical potentials in promoting wound healing. Method HuMSCs were isolated and expanded. When the cells of third passage reached confluency, ascorbic acid was added to stimulate the cells to deposit ECM where the cells grew in. Four weeks later, a cells-loaded ECM sheet was formed. The cell-ECM complex was observed under the scanning electron microscopy (SEM) and subjected to histological studies. The supernatants were collected and the cell-ECM complex was harvested at different time points and processed for enzyme-linked immune sorbent assay (ELISA) and mRNA analysis. The in vivo experiments were performed by means of implanting the cell-ECM complex on the mice back for up to 6 months and the specimens were collected for histological studies. Results After 4 weeks of cultivation with ascorbic stimulation, a sheet was formed which is mainly composed with HuMSCs, collagen and hyaluronic acid. The cell-ECM complex can sustain to certain tensile force. The mRNA and protein levels of vascular endothelial growth factor-α (VEGF-α), hepatocyte growth factor (HGF), keratinocyte growth factor (KGF), and transforming growth factor-β1 (TGF-β1) were remarkably increased compared to monolayer-cultured cells. The implanted cell-ECM complex on mice was still noticeable with host cells infiltration and vascularization on 6 months. Conclusion Our studies suggested that HuMSCs can be multi-cultivated through adding ascorbic stimulation and ECM containing collagen and hyaluronic acid were enriched around the cells which self-assembly formed a cell-ECM complex. Cell-ECM complex can improve growth factors secretion remarkably which means it may promote wound healing by paracrine.
... Tissue engineering utilizing scaffolds together with cell therapy opened a window into the treatment of burn injuries, resulting in faster and more effective wound healing [29]. An attractive strategy is to use biocompatible materials to replace inadequate healthy skin of the patients [30]. ...
Article
Full-text available
Background In vivo cell tracking after transplantation in regenerative medicine remains an unmet challenge and limits current understanding of the wound healing mechanism through cell-based therapies. This study investigated tracking of human Wharton’s jelly stem cells (hWJSCs) seeded onto an acellular dermal matrix (ADM) and labeled with superparamagnetic iron oxide nanoparticles (SPIONs) by magnetic resonance imaging (MRI) in burn injury. Method The hWJSCs were characterized and assessed for growth kinetics. A total of 30 rats were enrolled in three equal groups. Group 1 underwent scald burn injury left without treatment, the group 2 was treated by an ADM that was prepared from cosmetic surgery skin samples and the group 3 received hWJSCs labeled with SPIONs seeded onto an ADM. Tensile strength was evaluated before and after interventions, real time PCR assessed apoptosis, and Prussian blue staining, scanning electron microscopy (SEM) and MRI were used for the tracking of labeled cells. Results The hWJSCs exhibited mesenchymal stem cell properties. Population doubling time was 40.1 hours. SPIONs did not show any toxic effect. The hWJSCs seeded onto an ADM decreased Bax and increased Bcl-2 gene expression. Internalization of SPIONs within hWJSCs was confirmed by Prussian blue staining, SEM and MRI until day 21. There was a significant difference between the Young’s moduli of normal skin and the group receiving hWJSCs seeded onto an ADM. Histological observations and SEM imaging confirmed that MRI is an accurate method to track SPION-labeled hWJSCs in vivo. Conclusions This study showed that SPION labeling coupled with MRI can be used to further understand the fate of stem cells after transplantation in a burn model.
... Inducing angiogenesis leading to tissue repair [136] Excisional wound Porcine MSCs seeded on collagen Improving MSC adhesion and infiltration and supporting the wound healing [137] Excisional wound Mice BM-MSCs seeded on ADM Supported angiogenesis as well as collagen fiber structural remodeling [138] Excisional wound Mice MSCs seeded on fibrin hydrogels Stimulating endothelial cell proliferation, promoting macrophage polarization log with improving angiogenesis [139] Excisional wound Mice ISCs and MSCs coencapsulated into a synthetic hydrogel Higher healing response than singly delivered MSCs or ISCs [140] macrophage's inflammatory phenotype contributed to the inflammatory phase, potentiate new blood vessels generation, leading to the promoting angiogenesis, and finally facilitate construction of granulation tissues, skin cells, and ECM production [120]. ...
... Thereby, it was evidenced that such scaffolds could augment the capable presence of MSCs in wound sites to serve better therapeutic outcomes. Recently, transplantation of GFP-MSCs-seeded on an acellular dermal matrix (ADM) scaffolds into surgical skin wounds in vivo demonstrated that injected cells were retained inside the regenerating skin till 2 weeks upon transplantation [138]. Given the observations, injected MSCs differentiated into functional cells and enabled recruitment of more endogenous cells for tissue remodeling by secreting soluble mediators. ...
... Given the observations, injected MSCs differentiated into functional cells and enabled recruitment of more endogenous cells for tissue remodeling by secreting soluble mediators. Notwithstanding, further analysis indicated that endogenous cells, but not exogenous cells, convinced skin wound healing during the later stage [138]. Also, MSC spheroids in stiffer gels were found that could produce higher levels of pro-angiogenic factor VEGF and anti-inflammatory factor PGE2 than naive MSCs, stimulating endothelial cell proliferation, improving macrophage polarization, and accelerating angiogenesis in vivo [139]. ...
Article
Full-text available
Recently, mesenchymal stromal cells (MSCs) and also their exosome has become a game-changing tool in the context of tissue engineering and regenerative medicine. MSCs due to their competencies to establish skin cells, such as fibroblast and keratinocyte, and also their unique attribute to suppress inflammation in wound site has attracted increasing attention among scholars. In addition, MSC’s other capabilities to induce angiogenesis as a result of secretion of pro-angiogenic factors accompanied with marked anti-fibrotic activities, which mainly mediated by the releases matrix metalloproteinase (MMPs), make them a rational and effective strategy to accelerate wound healing with a small scar. Since the chief healing properties of the MSCs depend on their paracrine effects, it appears that MSCs-derived exosomes also can be an alternative option to support wound healing and skin regeneration as an innovative cell-free approach. Such exosomes convey functional cargos (e.g., growth factor, cytokine, miRNA, etc.) from MSCs to target cells, thereby affecting the recipient skin cells’ biological events, such as migration, proliferation, and also secretion of ECM components (e.g., collagen). The main superiorities of exosome therapy over parental MSCs are the diminished risk of tumor formation and also lower immunogenicity. Herein, we deliver an overview of recent in vivo reports rendering the therapeutic benefits of the MSCs-based therapies to ease skin wound healing, and so improving quality of life among patients suffering from such conditions.
... However, the roles of HIF1 in mitochondrial homeostasis and HaCaT survival under an oxidative microenvironment have not been adequately explained [33]. In addition, OPA1 is a mitochondrial protector in response to several stress responses, including inflammation stimulus [34], ischemia attack [35], and oxidative injury [36]. Activated OPA1 attenuates mitochondrial oxidative stress, sustains the mitochondrial membrane potential, and blocks the activation of mitochondrial apoptosis, thus sending a prosurvival signal to mitochondria [37][38][39]. ...
Article
Full-text available
Promoting epidermal cell survival in an oxidative stress microenvironment is vital for skin regeneration after burns and/or wounds. However, few studies have explored the mediators related to epidermal cell apoptosis in an oxidative stress microenvironment. Cellular viability was determined using the MTT assay, TUNEL staining, western blot analysis and LDH release assay. Two independent siRNAs were transfected into HaCaT cell to repress INF2 and/or HIF1 in the presence of H2O2. Mitochondrial function was determined using JC-1 staining, mitochondrial ROS staining, immunofluorescence staining and western blotting. In the present study, our data demonstrated that the expression of inverted formin-2 (INF2) increased rapidly when the cells were exposed to H2O2. Interestingly, INF2 knockdown promoted HaCaT cell survival via reducing H2O2-mediated cell apoptosis. Molecular investigations demonstrated that INF2 deletion attenuated mitochondrial ROS overloading, restored the cellular redox balance, sustained the mitochondrial membrane potential, improved mitochondrial respiratory function and corrected the mitochondrial dynamics disorder in an H2O2-mimicking oxidative stress microenvironment. In addition, INF2 deletion upregulated the expression of HIF1. Interestingly, the inhibition of HIF1 increased cell death and caused mitochondrial stress despite the deletion of INF2, suggesting that the HIF1 signaling pathway is required for INF2 deletion-mediated HaCaT cell survival and mitochondrial protection. Altogether, our results identified INF2 as a novel apoptotic mediator for oxidative stress-mediated HaCaT cell death via modulating mitochondrial stress and repressing the HIF1 signaling pathway. This finding provides evidence to support the critical role played by the INF2-HIF1 axis in regulating mitochondrial stress and epidermal cell viability in an oxidative stress microenvironment.
... Our study also found that Mst1 expression was much higher in the myocardium post-MI and that this contributed to post-infarction cardiac injury through the promotion of cardiomyocyte mitochondrial apoptosis. These data validate Mst1 as a vital regulator of cellular survival, highlighting that Mst1 may be a target for sustaining cardiomyocyte viability in response to acute and/or chronic stress damage [48,49]. ...
Article
Full-text available
Background Post-infarction cardiac injury is closely associated with cardiac remodeling and heart dysfunction. Mammalian STE20-like kinase 1 (Mst1), a regulator of cellular apoptosis, is involved in cardiac remodeling in post-infarction heart, but the mechanisms remain poorly defined. We aimed to explore the role of Mst1 in regulating chronic post-infarction cardiac injury, with a focus on mitochondrial homoeostasis. Methods Wild-type (WT) and Mst1-knockout mice were as the cardiac myocardial infarction model. Cardiac fibrosis, myocardial inflammation response, heart dysfunction and cardiomyocyte death were measured in vivo using immunohistochemistry, immunofluorescence, western blot, qPCR and TUNEL assays. Cardiomyocytes were isolated from WT and Mst1-knockout mice, and a chronic hypoxia model was used to induce damage. Mitochondrial function was determined via JC1 staining, ROS measurement, cyt-c leakage detection and mitochondrial apoptotic pathways analysis. Mitochondrial fission was observed using immunofluorescence. A pathway activator and inhibitor were applied to establish the signaling pathways involved in regulating mitochondrial homeostasis. Results Our study demonstrated that Mst1 expression was significantly upregulated in the heart post-infarction. Activated Mst1 induced cardiac fibrosis, an excessive inflammatory response, and cardiomyocyte death, whereas the genetic ablation of Mst1 protected the myocardium against chronic post-infarction injury. Function assays showed that upregulation of Mst1 activity contributed to JNK pathway activation, which led to Drp1 migration from the cytoplasm onto the surface of the mitochondria, indicative of mitochondrial fission activation. Excessive mitochondrial fission caused mitochondrial fragmentation, resulting in mitochondrial potential collapse, ROS overproduction, mitochondrial pro-apoptotic leakage into the cytoplasm, and the initiation of caspase-9-mediated mitochondrial apoptosis. By contrast, Mst1 deletion helped to maintain mitochondrial structure and function, sending pro-survival signals to the cardiomyocytes. Conclusions Our results identify Mst1 as a malefactor in the development of post-infarction cardiac injury and that it acts through the JNK-Drp1-mitochondrial fission pathway.
... However, these authors argued that activated NR4A1 due to high-fat diets mediates Drp1 phosphorylation and represses Bnip3 transcription, leading to fission activation and mitophagy inhibition. Other signaling pathways may also be involved in simultaneously regulating fission and mitophagy [70,71]. In cardiac microvascular ischemia reperfusion, DUSP1 has been shown to repress fission and recuse mitophagy via the JNK pathway [17,72]. ...
Article
Full-text available
Background/aims: Disrupted mitochondrial dynamics, including excessive mitochondrial fission and mitophagy arrest, has been identified as a pathogenic factor in diabetic nephropathy (DN), although the upstream regulatory signal for mitochondrial fission activation and mitophagy arrest in the setting of DN remains unknown. Methods: Wild-type (WT) mice and NR4A1 knockout (NR4A1-KO) mice were used to establish a DN model. Mitochondrial fission and mitophagy were evaluated by western blotting and immunofluorescence. Mitochondrial function was assessed by JC-1 staining, the mPTP opening assay, immunofluorescence and western blotting. Renal histopathology and morphometric analyses were conducted via H&E, Masson and PASM staining. Kidney function was evaluated via ELISA, western blotting and qPCR. Results: In the present study, we found that nuclear receptor subfamily 4 group A member 1 (NR4A1) was actually activated by a chronic hyperglycemic stimulus. Higher NR4A1 expression was associated with glucose metabolism disorder, renal dysfunction, kidney hypertrophy, renal fibrosis, and glomerular apoptosis. At the molecular level, increased NR4A1 expression activated p53, and the latter selectively stimulated mitochondrial fission and inhibited mitophagy by modulating Mff and Parkin transcription. Excessive Mff-related mitochondrial fission caused mitochondrial oxidative stress, promoted mPTP opening, exacerbated proapoptotic protein leakage into the cytoplasm, and finally initiated mitochondria-dependent cellular apoptosis in the setting of diabetes. In addition, defective Parkin-mediated mitophagy repressed cellular ATP production and failed to correct the uncontrolled mitochondrial fission. However, NR4A1 knockdown interrupted the Mff-related mitochondrial fission and recused Parkin-mediated mitophagy, reducing the hyperglycemia-mediated mitochondrial damage and thus improving renal function. Conclusion: Overall, we have shown that NR4A1 functions as a novel malefactor in diabetic renal damage and operates by synchronously enhancing Mff-related mitochondrial fission and repressing Parkin-mediated mitophagy. Thus, finding strategies to regulate the balance of the NR4A1-p53 signaling pathway and mitochondrial homeostasis may be a therapeutic option for treating diabetic nephropathy in clinical practice.
... To investigate cellular apoptosis, TUNEL assays and trypan blue staining were performed. A TUNEL assay was performed using a TUNEL assay kit (Roche Applied Science, Madison, WI, USA) according to the manufacturer's protocol (42). Images were captured using an inverted microscope (magnification, x40; BX51; Olympus Corporation). ...
Article
Mitophagy is important for cancer development. Notably, the role of Parkin‑mediated mitophagy in colorectal cancer (CRC) mortality has not been fully determined. Therefore, the present study aimed to investigate the effect of Parkin‑mediated mitophagy on CRC apoptosis. In addition, the present study investigated the therapeutic effects of Tanshinone IIA (Tan IIA) on the regulation of CRC cell death via mitophagy. Cellular apoptosis was measured following Tan IIA treatment. In addition, mitophagy activity was evaluated by immunofluorescence and western blotting. The results of the present study revealed that Tan IIA may enhance CRC cell death. In addition, the results demonstrated that Tan IIA enhanced mitochondrial apoptosis, as demonstrated by reduced mitochondrial membrane potential, elevated mitochondrial permeability transition pore opening, and increased oxidative stress, mitochondrial energy disorder and proapoptotic factor expression. Furthermore, the results of the present study demonstrated that Tan IIA induced mitochondrial apoptosis via inhibition of mitophagy. In addition, it was revealed that mitophagy could suppress mitochondrial apoptosis. Functional assays revealed that Tan IIA suppressed the adenosine monophosphate‑activated protein kinase (AMPK) pathway, resulting in the inactivation of S‑phase kinase associated protein 2 (Skp2). Furthermore, reduced levels of Skp2 failed to activate Parkin, thus resulting in inhibition of mitophagy. Conversely, reactivation of AMPK and overexpression of Skp2 rescued mitophagy activity and thus attenuated the Tan IIA‑induced apoptosis of CRC cells. In conclusion, the results of the present study demonstrated the beneficial role of mitophagy in CRC cell survival and suggested that Tan IIA may be an effective therapeutic agent, which suppresses mitophagy activity and enhances CRC apoptosis.
... The experiments were repeated three times with triplicates of each sample. Fold change of Mst1 and Parkin mRNA expression was normalized by GAPDH as an internal control [40]. ...
Article
Full-text available
Background/aims: Mitochondrial homeostasis is implicated in the development and progression of endometriosis through poorly defined mechanisms. Mst1 is the major growth suppressor related to cancer migration, apoptosis and proliferation. However, whether Mst1 is involved in endometriosis apoptosis and migration via regulating the mitochondrial function remains to be elucidated. Methods: Expression of Mst1 in endometriosis was examined via western blots. Cellular apoptosis was detected via MTT and TUNEL assay. Gain of function assay about Mst1 was conducted via adenovirus over-expression. Mitochondrial functions were evaluated via mitochondrial membrane potential JC-1 staining, ROS flow cytometry analysis, mPTP opening assessment and immunofluorescence of HtrA2/Omi. The mitophagy activity were examined via western blots and immunofluorescence. Results: First, we found that Mst1 was significantly downregulated in the ectopic endometrium of endometriosis compared to the normal endometrium. However, the recovery of Mst1 function was closely associated with the inability of endometrial stromal cells (ESCs) to migrate and survive. A functional study indicated that regaining Mst1 enhanced Drp1 post-transcriptional phosphorylation at Ser616 and repressed Parkin transcription activity via p53, leading to mitochondrial fission activation and mitophagy inhibition. Excessive Drp1-related fission forced the mitochondria to liberate HtrA2/Omi into the cytoplasm. Moreover, Mst1-induced defective mitophagy evoked cellular oxidative stress, energy metabolism and calcium overload. Through excessive mitochondrial fission and aberrant mitophagy, Mst1 launched caspase 9-related mitochondrial apoptosis and abrogated F-actin/lamellipodium-dependent cellular migration. Notably, we also defined NR4A/miR181c as the upstream signal for Mst1 dysfunction in endometriosis. Conclusion: Collectively, our results comprehensively described the important role of the NR4A-miR181c-Mst1 pathway in endometriosis, which handled mitochondrial apoptosis and F-actin/ lamellipodium-based migration via the regulation of Drp1-related mitochondrial fission and Parkin-required mitophagy, with a potential application in endometriosis therapy by limiting ESCs migration and promoting apoptosis.
... GFP + BMSCs were obtained as previously reported by our group [30]. BMSC sheets were prepared by plating 1.5 × 10 5 of third passage cells on culture dishes (six-well plates, Corning). ...
... More BMSCs survived and, of these, more BMSCs were in the cell replication period of S, G2, or M phase after treatment with curcumin, as shown in Fig. 2f, suggesting that curcumin significantly enhanced the proliferation of BMSCs. Previous research by ourselves and others have shown that BMSCs affect other cells primarily through paracrine effects to participate in the process of tissue repair and immune regulation [30]. Greater numbers and longer survival rates ensured that the BMSCs secreted more cytokines to promote wound healing by enhancing the immunomodulatory ability of the above-described BMSCs. ...
Article
Full-text available
Background: Adult full-thickness cutaneous wound repair suffers from an imbalanced immune response, leading to nonfunctional reconstructed tissue and fibrosis. Although various treatments have been reported, the immune-mediated tissue regeneration driven by biomaterial offers an attractive regenerative strategy for damaged tissue repair. Methods: In this research, we investigated a specific bone marrow-derived mesenchymal stem cell (BMSC) sheet that was induced by the Traditional Chinese Medicine curcumin (CS-C) and its immunomodulatory effects on wound repair. Comparisons were made with the BMSC sheet induced without curcumin (CS-N) and control (saline). Results: In vitro cultured BMSC sheets (CS-C) showed that curcumin promoted the proliferation of BMSCs and modified the features of produced extracellular matrix (ECM) secreted by BMSCs, especially the contents of ECM structural proteins such as fibronectin (FN) and collagen I and III, as well as the ratio of collagen III/I. Two-photon fluorescence (TPF) and second-harmonic generation (SHG) imaging of mouse implantation revealed superior engraftment of BMSCs, maintained for 35 days in the CS-C group. Most importantly, CS-C created a favorable immune microenvironment. The chemokine stromal cell-derived factor 1 (SDF1) was abundantly produced by CS-C, thus facilitating a mass migration of leukocytes from which significantly increased expression of signature TH1 cells (interferon gamma) and M1 macrophages (tumor necrosis factor alpha) genes were confirmed at 7 days post-operation. The number of TH1 cells and associated pro-inflammatory M1 macrophages subsequently decreased sharply after 14 days post-operation, suggesting a rapid type I immune regression. Furthermore, the CS-C group showed an increased trend towards M2 macrophage polarization in the early phase. CS-C led to an epidermal thickness and collagen deposition that was closer to that of normal skin. Conclusions: Curcumin has a good regulatory effect on BMSCs and this promising CS-C biomaterial creates a pro-regenerative immune microenvironment for cutaneous wound healing.
... Notably, previous studies have reported that the actin cytoskeleton is regulated by p53 via activation of HtrA2/Omi [56], which hampers cell invasion. However, our studies further characterized the mechanism by which HtrA2/Omi disturbs actin homeostasis [57]. ...
Article
Full-text available
Background/aims: The Hippo-Yap pathway is associated with tumor development and progression. However, little evidence is available concerning its role in cancer cell apoptosis and migration via mitochondrial homeostasis. Here, we identify mitochondrial fission as a regulator of the Hippo-Yap pathway in human rectal cancer tumorigenesis and metastasis. Methods: In this study, we performed loss-of function assays concerning Yap in RCC via shRNA. Cellular viability and apoptosis were measured via MTT, the TUNEL assay and trypan blue staining. Mitochondrial function was assessed via JC1 staining, the mPTP opening assay, mitochondrial respiratory function analysis, electron microscopy and immunofluorescence analysis of HtrA2/Omi. Mitophagy and mitochondrial fission were assessed via western blots and immunofluorescence. Cell migration was evaluated via the Transwell assay, wound-healing assay and immunofluorescence analysis of F-actin. The interaction between JNK and Yap was detected via co-immunoprecipitation and Yap recombinant mutagenic plasmid transfection. Western blots were used to analyze signaling pathways in conjunction with JNK inhibitors or HtrA2/Omi siRNA. Results: Yap is upregulated in human rectal cancer cells, where its expression correlates positively with cell survival and migration. Functional studies established that silencing of Yap drove JNK phosphorylation, which induced Drp1 activation and translocation to the surface of mitochondria, initiating mitochondrial fission. Excessive mitochondrial fission mediated HtrA2/Omi leakage from the mitochondria into the cytoplasm, where HtrA2/Omi triggered cellular apoptosis via the mitochondrial apoptosis pathway. Moreover, released HtrA2/Omi also phosphorylated cofilin and inhibited cofilin-mediated F-actin polymerization. F-actin collapse perturbed lamellipodia formation and therefore impaired cellular migration and invasion. Conclusion: Collectively, our results demonstrate that Hippo-Yap can serve as a tumor promoter in human rectal cancer and acts by restricting JNK/Drp1/mitochondrial fission/ HtrA2/Omi, with potential implications for new approaches to human rectal cancer therapy.