Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 176: 116848, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38834005

ABSTRACT

Liver fibrosis is an intrahepatic chronic damage repair response caused by various reasons such as alcoholic liver, fatty liver, viral hepatitis, autoimmune diseases, etc., and is closely related to the progression of liver disease. Currently, the mechanisms of liver fibrosis and its treatment are hot research topics in the field of liver disease remedy. Mesenchymal stem cells (MSCs) are a class of adult stem cells with self-renewal and multidirectional differentiation potential, which can ameliorate fibrosis through hepatic-directed differentiation, paracrine effects, and immunomodulation. However, the low inner-liver colonization rate, low survival rate, and short duration of intervention after stem cell transplantation have limited their wide clinical application. With the intensive research on liver fibrosis worldwide, it has been found that MSCs and MSCs-derived exosomes combined with drugs have shown better intervention efficiency than utilization of MSCs alone in many animal models of liver fibrosis. In this paper, we review the interventional effects and mechanisms of mesenchymal stem cells and their exosomes combined with drugs to alleviate hepatic fibrosis in vivo in animal models in recent years, which will provide new ideas to improve the efficacy of mesenchymal stem cells and their exosomes in treating hepatic fibrosis in the clinic.

2.
Inflammation ; 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441793

ABSTRACT

Psoriasis is a prevalent condition characterized by chronic inflammation, immune dysregulation, and genetic alterations, significantly impacting the well-being of affected individuals. Recently, a novel aspect of programmed cell death, ferroptosis, linked to iron metabolism, has come to light. This research endeavors to unveil novel diagnostic genes associated with ferroptosis in psoriasis, employing bioinformatic methods and experimental validation. Diverse analytical strategies, including "limma," Weighted Gene Co-expression Network Analysis (WGCNA), Least Absolute Shrinkage and Selection Operator (LASSO), Support Vector Machine Recursive Feature Elimination (SVM-RFE), and Random Forest (RF), were employed to pinpoint pivotal ferroptosis-related diagnostic genes (FRDGs) in the training datasets GSE30999, testing dataset GSE41662 and GSE14905. The discriminative potential of FRDGs in distinguishing between normal and psoriatic patients was gauged using Receiver Operating Characteristic (ROC) curves, while the functional pathways of FRDGs were scrutinized through Gene Set Enrichment Analysis (GSEA). Spearman correlation and ssGSEA analysis were applied to explore correlations between FRDGs and immune cell infiltration or oxidative stress-related pathways. The study identified six robust FRDGs - PPARD, MAPK14, PARP9, POR, CDCA3, and PDK4 - which collectively formed a model boasting an exceptional AUC value of 0.994. GSEA analysis uncovered their active involvement in psoriasis-related pathways, and substantial correlations with immune cells and oxidative stress were noted. In vivo, experiments confirmed the consistency of the six FRDGs in the psoriasis model with microarray results. In vitro, genetic knockdown or inhibition of MAPK14 using SW203580 in keratinocytes attenuated ferroptosis and reduced the expression of inflammatory cytokines. Furthermore, the study revealed that intercellular communication between keratinocytes and macrophages was augmented by ferroptotic keratinocytes, increased M1 polarization, and recruitment of macrophage was regulated by MAPK14. In summary, our findings unveil novel ferroptosis-related targets and enhance the understanding of inflammatory responses in psoriasis. Targeting MAPK14 signaling in keratinocytes emerges as a promising therapeutic approach for managing psoriasis.

3.
Stem Cells Transl Med ; 13(2): 151-165, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-37936499

ABSTRACT

Hepatocyte-like cells (HLCs) that are differentiated from mesenchymal stem cells (MSCs) provide a valuable resource for drug screening and cell-based regeneration therapy. Differentiating HLCs into 3D spheroids enhances their phenotypes and functions. However, the molecular mechanisms underlying MSCs hepatogenic differentiation are not fully understood. In this study, we generated HLCs from human adipose-derived mesenchymal stem cells (hADMSCs) in both 2D and 3D cultures. We performed an acetyl-proteomics assay on the HLCs derived from both 2D and 3D differentiation and identified a differential change in H3K56 acetylation between the 2 differentiated cells. Our findings revealed that 3D differentiation activated ALB gene transcription by increasing the acetylation level of H3K56, thereby enhancing the phenotypes and functions of HLCs and further promoting their maturation. Notably, inhibiting p300 reduced the acetylation level of H3K56 during hepatogenic differentiation, leading to decreased phenotypes and functions of HLCs, whereas activation of p300 promoted hepatogenic differentiation, suggesting that p300 plays a critical role in this process. In summary, our study demonstrates a potential mechanism through which 3D spheroids differentiation facilitates hADMSCs differentiation into HLCs by promoting p300-mediated H3K56 acetylation, which could have significant clinical applications in liver regeneration and disease modeling.


Subject(s)
Hepatocytes , Mesenchymal Stem Cells , Humans , Acetylation , Cell Differentiation , Cells, Cultured
4.
Cell Transplant ; 32: 9636897231210069, 2023.
Article in English | MEDLINE | ID: mdl-37982384

ABSTRACT

Human umbilical cord mesenchymal stem cells (hUCMSC) have shown promising potential in ameliorating brain injury, but the mechanism is unclear. We explore the role of NogoA/NgR/Rho pathway in mediating hUCMSC to improve neurobehavioral status and alleviate brain injury in hypoxia/ischemia-induced CP (cerebral palsy) rat model in order to promote the clinical application of stem cell therapy in CP. The injury model of HT22 cells was established after 3 h hypoxia, and then co-cultured with hUCMSC. The rat model of CP was established by ligation of the left common carotid artery for 2.5 h. Subsequently, hUCMSC was administered via the tail vein once a week for a total of four times. The neurobehavioral status of CP rats was determined by behavioral experiment, and the pathological brain injury was determined by pathological staining method. The mRNA and protein expressions of NogoA, NgR, RhoA, Rac1, and CDC42 in brain tissues of rats in all groups and cell groups were detected by real-time quantitative polymerase chain reaction (RT-qPCR), Western blot, and immunofluorescence. The CP rats exhibited obvious motor function abnormalities and pathological damage. Compared with the control group, hUCMSC transplantation could significantly improve the neurobehavioral situation and attenuate brain pathological injury in CP rats. The relative expression of NogoA, NgR, RhoA mRNA, and protein in brain tissues of rats in the CP group was significantly higher than the rats in the sham and CP+hUCMSC group. The relative expression of Rac1, CDC42 mRNA, and protein in brain tissues of rats in the CP group was significantly lower than the rats in the sham and CP+hUCMSC group. The animal experiment results were consistent with the experimental trend of hypoxic injury of HT22 cells. This study confirmed that hUCMSC can efficiently improve neurobehavioral status and alleviate brain injury in hypoxia/ischemia-induced CP rat model and HT22 cell model through downregulating the NogoA/NgR/Rho pathway.


Subject(s)
Brain Injuries , Cerebral Palsy , Mesenchymal Stem Cells , Rats , Humans , Animals , Hypoxia/metabolism , Ischemia/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism , Brain Injuries/metabolism , RNA, Messenger/metabolism
5.
Stem Cell Res Ther ; 14(1): 301, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37864199

ABSTRACT

AIM: Although the efficacy and safety of mesenchymal stem cell therapy for liver cirrhosis have been demonstrated in several studies. Clinical cases of mesenchymal stem cell therapy for patients with liver cirrhosis are limited and these studies lack the consistency of treatment effects. This article aimed to systematically investigate the efficacy and safety of mesenchymal stem cells in the treatment of liver cirrhosis. METHOD: The data source included PubMed/Medline, Web of Science, EMBASE, and Cochrane Library, from inception to May 2023. Literature was screened by the PICOS principle, followed by literature quality evaluation to assess the risk of bias. Finally, the data from each study's outcome indicators were extracted for a combined analysis. Outcome indicators of the assessment included liver functions and adverse events. Statistical analysis was performed using Review Manager 5.4. RESULTS: A total of 11 clinical trials met the selection criteria. The pooled analysis' findings demonstrated that both primary and secondary indicators had improved. Compared to the control group, infusion of mesenchymal stem cells significantly increased ALB levels in 2 weeks, 1 month, 3 months, and 6 months, and significantly decreased MELD score in 1 month, 2 months, and 6 months, according to a subgroup analysis using a random-effects model. Additionally, the hepatic arterial injection favored improvements in MELD score and ALB levels. Importantly, none of the included studies indicated any severe adverse effects. CONCLUSION: The results showed that mesenchymal stem cell was effective and safe in the treatment of liver cirrhosis, improving liver function (such as a decrease in MELD score and an increase in ALB levels) in patients with liver cirrhosis and exerting protective effects on complications of liver cirrhosis and the incidence of hepatocellular carcinoma. Although the results of the subgroup analysis were informative for the selection of mesenchymal stem cells for clinical treatment, a large number of high-quality randomized controlled trials validations are still needed.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mesenchymal Stem Cell Transplantation , Humans , Mesenchymal Stem Cell Transplantation/adverse effects , Liver Cirrhosis/therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology
6.
Toxicology ; 492: 153532, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37141935

ABSTRACT

Thallium (Tl) is a high-priority toxic metal that poses a severe threat to human health. The toxicity characteristics induced by Tl have been partially discussed. However, the immunotoxic effects of Tl exposure have remained largely unexplored. Our findings demonstrated that 50 ppm of Tl exposure for one week induced severe weight loss in mice, which was accompanied by appetite suppression. Moreover, although Tl exposure did not induce significant pathological damage to skeletal muscle and bone, Tl inhibited the expression of B cell development-related genes in the bone marrow. Additionally, Tl exposure increased B cell apoptosis and reduced its generation in the bone marrow. Analysis of B cells in the blood indicated that the percentage of B-2 cells decreased significantly, whereas B-2 cell proportions in the spleen did not. The percentage of CD4+ T cells in the thymus increased significantly, and the proportion of CD8+ T cells did not. Furthermore, although the proportion of the total CD4+ and CD8+ T cells was not significantly altered in the blood and spleen, Tl exposure promoted the migration of naïve CD4+ T cells and recent thymic emigrants (RTEs) from the thymus to the spleen. These results suggest that Tl exposure can affect B and T cell generation and migration, which provides new evidence for Tl-induced immunotoxicity.


Subject(s)
B-Lymphocytes , T-Lymphocytes , Thallium , Thallium/toxicity , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , T-Lymphocytes/drug effects , Animals , Mice , Cell Movement/drug effects , Gene Expression/drug effects , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Thymus Gland/cytology , Thymus Gland/drug effects , Bone Marrow/drug effects , Body Weight/drug effects
7.
Front Pediatr ; 10: 897398, 2022.
Article in English | MEDLINE | ID: mdl-35601435

ABSTRACT

Aim: There is insufficient evidence regarding the efficacy and safety of stem cell therapy for autism spectrum disorders. We performed the first meta-analysis of stem cell therapy for autism spectrum disorders in children to provide evidence for clinical rehabilitation. Methods: The data source includes PubMed/Medline, Web of Science, EMBASE, Cochrane Library and China Academic Journal, from inception to 24th JULY 2021. After sifting through the literature, the Cochrane tool was applied to assess the risk of bias. Finally, we extracted data from these studies and calculated pooled efficacy and safety. Results: 5 studies that met the inclusion criteria were included in current analysis. Meta-analysis was performed using rehabilitation therapy as the reference standard. Data showed that the Childhood Autism Rating Scale score of stem cell group was striking lower than the control group (WMD: -5.96; 95%CI [-8.87, -3.06]; p < 0.0001). The Clinical Global Impression score consolidated effect size RR = 1.01, 95%CI [0.87, 1.18], Z = 0.14 (p = 0.89), the effective rate for The Clinical Global Impression was 62% and 60% in the stem cell group and the control group, respectively. The occurrence events of adverse reactions in each group (RR = 1.55; 95%CI = 0.60 to 3.98; p = 0.36), there was no significant difference in the incidence of adverse reactions between the stem cell group and the control group. Conclusions: The results of this meta-analysis suggested that stem cell therapy for children with autism might be safe and effective. However, the evidence was compromised by the limitations in current study size, lacking standardized injection routes and doses of stem cells, as well as shortages in diagnostic tools and long period follow-up studies. Hence, it calls for more studies to systematically confirm the efficacy and safety of stem cell therapy for children with autism spectrum disorders.

8.
Biomed Res Int ; 2022: 6484073, 2022.
Article in English | MEDLINE | ID: mdl-35572725

ABSTRACT

Human tissue-plasminogen activator (tPA) is a thrombolytic drug widely used in the treatment of stroke, pulmonary thrombosis, acute myocardial infarction, and other thrombotic diseases. The double genes cointegrated into the organisms and cells can produce a synergistic effect, which will improve the expression level of the target gene. However, the study of the integration of the GH and tPA genes to improve the expression level of tPA has not yet been reported. In order to elucidate this, we generated monoclonal goat mammary epithelial cell lines with tPA/GH double-gene integration and analyzed the tPA expression level in single- and double-gene integrated cells. We selected the mammary gland-specific expressing vectors BLC14/tPA and BLC14/GH with the ß-lactoglobulin gene as a regulatory sequence in our previous research. The tPA and GH genes were electronically cotransfected into goat mammary epithelial cells. Resistant cell lines were screened by G418, and transgenic monoclonal cell lines were confirmed by PCR. The tPA expression was induced by prolactin and detected in the cell induction solution after 48 h by ELISA and Western blotting. We detected the tPA biological activity in vitro by fibrin agarose plate assay (FAPA). The results showed that a total of 207 resistant monoclonal cells were obtained, including 126 cell lines with tPA monogenic integration and 51 cell lines with tPA/GH double-gene integration. The rate of double-gene integration was 24.6% (51/207). A total of 48 cells expressed tPA, of which 25.3% (19/75) cells expressed single gene, and 56.9% (29/51) cells expressed double genes. The concentration of tPA in single-gene-expressing cells was 8.0-64.0 µg/mL, and the tPA level in double-gene-expressing cells was significantly higher (200-7200 µg/mL). In addition, the tPA had a relatively strong in vitro thrombolytic activity determined by FAPA. The results showed that goat mammary epithelial cell lines with tPA/GH gene integration were successfully established by electrotransfection, and the expression level of tPA in double-gene integrated cell lines was significantly increased. This study provided a new way for the preparation of a transgenic goat and other animal with high tPA expression by somatic cell nuclear transfer. The findings also laid a foundation for efficient production of pharmaceutical proteins in transgenic animal mammary gland bioreactors in the future.


Subject(s)
Fibrinolytic Agents , Goats , Animals , Animals, Genetically Modified , Epithelial Cells , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Goats/genetics , Mammary Glands, Animal/metabolism
9.
ACS Appl Mater Interfaces ; 14(22): 25155-25172, 2022 Jun 08.
Article in English | MEDLINE | ID: mdl-35618676

ABSTRACT

The repair of spinal cord injury (SCI) is still a tough clinical challenge and needs innovative therapies. Mitochondrial function is significantly compromised after SCI and has emerged as an important factor causing neuronal apoptosis and hindering functional recovery. In this study, umbilical cord mesenchymal stem cells (UCMSC), which are promising seed cells for nerve regeneration, and basic fibroblast growth factor (bFGF) that have been demonstrated to have a variety of effects on neural regeneration were jointly immobilized in extracellular matrix (ECM) and heparin-poloxamer (HP) to create a polymer bioactive system that brings more hope and possibility for the treatment of SCI. Our results in vitro and in vivo showed that the UCMSC-bFGF-ECM-HP thermosensitive hydrogel has good therapeutic effects, mainly in reducing apoptosis and improving the mitochondrial function. It showed promising utility for the functional recovery of impaired mitochondrial function by promoting mitochondrial fusion, reducing pathological mitochondrial fragmentation, increasing mitochondrial energy supply, and improving the metabolism of MDA, LDH, and ROS. In addition, we uncovered a distinct molecular mechanism underlying the protective effects associated with activating p21-activated kinase 1 (PAK1) and mitochondrial sirtuin 4 (SIRT4) by the UCMSC-bFGF-ECM-HP hydrogel. The expansion of new insights into the molecular relationships between PAK1 and SIRT4, which links the mitochondrial function in SCI, can lay the foundation for future applications and help to provide promising interventions of stem-cell-based biological scaffold therapies and potential therapeutic targets for the clinical formulation of SCI treatment strategies.


Subject(s)
Spinal Cord Injuries , Spinal Cord Regeneration , Animals , Heparin/therapeutic use , Hydrogels/pharmacology , Hydrogels/therapeutic use , Mitochondria/metabolism , Poloxamer/therapeutic use , Rats , Rats, Sprague-Dawley
10.
ACS Omega ; 7(4): 3738-3745, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35128282

ABSTRACT

Liver fibrosis is the intermediate process and inevitable stage of the development of chronic liver disease into cirrhosis. Reducing the degree of liver fibrosis plays an extremely important role in treating chronic liver disease and preventing liver cirrhosis and liver cancer. The formation of liver fibrosis is affected by iron deposition to a certain extent, and excessive iron deposition further induces liver cirrhosis and liver cancer. Herein, confocal microbeam X-ray fluorescence (µ-XRF) was used to determine the intensity and biodistribution of iron deposition at different time points in the process of liver fibrosis induced by thioacetamide (TAA) in rats. To our best knowledge, this is the first study using confocal µ-XRF to analyze hepatic iron deposition in hepatic fibrosis. The results showed that there are minor and trace elements such as iron, potassium, and zinc in the liver of rats. Continuous injection of TAA solution resulted in increasing liver iron deposition over time. The intensity of iron deposition in liver tissue was also significantly reduced after bone mesenchymal stem cells (BMSCs) were injected. These findings indicated that confocal µ-XRF can be used as a nondestructive and quantitative method of evaluating hepatic iron deposition in hepatic fibrosis, and iron deposition may play an important role in the progression of hepatic fibrosis induced by TAA.

11.
Front Bioeng Biotechnol ; 10: 1006845, 2022.
Article in English | MEDLINE | ID: mdl-36588957

ABSTRACT

Aim: Although the efficacy and safety of stem cell therapy for cerebral palsy has been demonstrated in previous studies, the number of studies is limited and the treatment protocols of these studies lack consistency. Therefore, we included all relevant studies to date to explore factors that might influence the effectiveness of treatment based on the determination of safety and efficacy. Methods: The data source includes PubMed/Medline, Web of Science, EMBASE, Cochrane Library, from inception to 2 January 2022. Literature was screened according to the PICOS principle, followed by literature quality evaluation to assess the risk of bias. Finally, the outcome indicators of each study were extracted for combined analysis. Results: 9 studies were included in the current analysis. The results of the pooled analysis showed that the improvements in both primary and secondary indicators except for Bayley Scales of Infant and Toddler Development were more skewed towards stem cell therapy than the control group. In the subgroup analysis, the results showed that stem cell therapy significantly increased Gross Motor Function Measure (GMFM) scores of 3, 6, and 12 months. Besides, improvements in GMFM scores were more skewed toward umbilical cord mesenchymal stem cells, low dose, and intrathecal injection. Importantly, there was no significant difference in the adverse events (RR = 1.13; 95% CI = [0.90, 1.42]) between the stem cell group and the control group. Conclusion: The results suggested that stem cell therapy for cerebral palsy was safe and effective. Although the subgroup analysis results presented guiding significance in the selection of clinical protocols for stem cell therapy, high-quality RCTs validations are still needed.

12.
Curr Neurovasc Res ; 18(1): 20-77, 2021.
Article in English | MEDLINE | ID: mdl-33745435

ABSTRACT

BACKGROUND: Previous studies have reported that mesenchymal stem cell (MSC)- derived exosomes can protect primary rat brain microvascular endothelial cells (BMECs) against oxygen-glucose deprivation and reoxygenation (OGD/R)-induced injury. OBJECTIVE: The aim was to identify the key factors mediating the protective effects of MSC-derived exosomes. METHODS: Primary rat BMECs were either pretreated or not pretreated with MSC-derived exosomes before exposure to OGD/R. Naïve cells were used as a control. After performing small RNA deep sequencing, quantitative reverse transcription polymerase chain reaction was performed to validate microRNA (miRNA) expression. The effects of rno-miR-666-3p on cell viability, apoptosis, and inflammation in OGD/R-exposed cells were assessed by performing the Cell Counting Kit 8 assay, flow cytometry, and enzyme-linked immunosorbent assay, respectively. Moreover, the role of rno-miR-666-3p in regulating gene expression in OGD/R-exposed cells was studied using mRNA deep sequencing. Lastly, to evaluate whether mitogen-activated protein kinase 1 (MAPK1) was the target of rno-miR-666-3p, western blotting and the dual-luciferase assay were performed. RESULTS: MSC-derived exosomes altered the miRNA expression patterns in OGD/R-exposed BMECs. In particular, the expression levels of rno-miR-666-3p, rno-miR-92a-2-5p, and rnomiR- 219a-2-3p decreased in OGD/R-exposed cells compared with those in the control; however, MSC-derived exosomes restored the expression levels of these miRNAs under OGD/R conditions. rno-miR-666-3p overexpression enhanced cell viability and alleviated the apoptosis of OGD/R-exposed cells. Moreover, rno-miR-666-3p suppressed OGD/R-induced inflammation. mRNA deep sequencing revealed that rno-miR-666-3p is closely associated with the MAPK signaling pathway. Western blotting and the dual-luciferase assay confirmed that MAPK1 is the target of rnomiR- 666-3p. CONCLUSION: MSC-derived exosomes restore rno-miR-666-3p expression in OGD/R-exposed BMECs. Moreover, this specific miRNA exerts protective effects against OGD/R by suppressing the MAPK signaling pathway.


Subject(s)
Brain/metabolism , Cell Survival/physiology , Endothelial Cells/metabolism , Exosomes/metabolism , MAP Kinase Signaling System/physiology , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Animals , Cell Hypoxia/physiology , Glucose/metabolism , Oxygen/metabolism , Rats
13.
Curr Neurovasc Res ; 17(2): 155-163, 2020.
Article in English | MEDLINE | ID: mdl-32056526

ABSTRACT

OBJECTIVE: The effects of mesenchymal stem cell (MSC)-derived exosomes on brain microvascular endothelial cells under oxygen-glucose deprivation (OGD), which mimic cells in deep hypothermic circulatory arrest (DHCA) in vitro, are yet to be studied. METHODS: MSCs were co-cultured with primary rat brain endothelial cells, which were then exposed to OGD. Cell viability, apoptosis, the inflammatory factors (IL-1ß, IL-6, and TNF-α), and the activation of inflammation-associated TLR4-mediated pyroptosis and the NF-κB signaling pathway were determined. Furthermore, exosomes derived from MSCs were isolated and incubated with endothelial cells to investigate whether the effect of MSCs is associated with MSCderived exosomes. Apoptosis, cell viability, and the inflammatory response were also analyzed in OGD-induced endothelial cells incubated with MSC-derived exosomes. RESULTS: OGD treatment promoted endothelial cell apoptosis, induced the release of inflammatory factors IL-1ß, IL-6, and TNF-α, and inhibited cell viability. Western blot analysis showed that OGD treatment-induced TLR4, and NF-κB p65 subunit phosphorylation and caspase-1 upregulation, while co-culture with MSCs could reduce the effect of OGD treatment on endothelial cells. As expected, the effect of MSC-derived exosomes on OGD-treated endothelial cells was similar to that of MSCs. MSC-derived exosomes alleviated the OGD-induced decrease in the viability of endothelial cells, and increased levels of apoptosis, inflammatory factors, and the activation of inflammatory and inflammatory focal pathways. CONCLUSION: Both MSCs and MSC-derived exosomes attenuated OGD-induced rat primary brain endothelial cell injury. These findings suggest that MSC-derived exosomes mediate at least some of the protective effects of MSCs on endothelial cells.


Subject(s)
Brain/metabolism , Cell Hypoxia/physiology , Endothelial Cells/metabolism , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis/physiology , Brain/cytology , Cell Survival/physiology , Coculture Techniques , Cytokines/metabolism , Endothelial Cells/cytology , Glucose/metabolism , Mesenchymal Stem Cells/cytology , NF-kappa B/metabolism , Oxygen/metabolism , Rats , Signal Transduction/physiology
14.
Biomed Mater Eng ; 28(s1): S113-S119, 2017.
Article in English | MEDLINE | ID: mdl-28372286

ABSTRACT

Liver transplantation is the definitive treatment for patients with end-stage liver diseases (ESLD). However, it is hampered by shortage of liver donor. Liver tissue engineering, aiming at fabricating new livers in vitro, provides a potential resolution for donor shortage. Three elements need to be considered in liver tissue engineering: seeding cell resources, scaffolds and bioreactors. Studies have shown potential cell sources as hepatocytes, hepatic cell line, mesenchymal stem cells and others. They need scaffolds with perfect biocompatiblity, suitable micro-structure and appropriate degradation rate, which are essential charateristics for cell attachment, proliferation and secretion in forming extracellular matrix. The most promising scaffolds in research include decellularized whole liver, collagens and biocompatible plastic. The development and function of cells in scaffold need a microenvironment which can provide them with oxygen, nutrition, growth factors, et al. Bioreactor is expected to fulfill these requirements by mimicking the living condition in vivo. Although there is great progress in these three domains, a large gap stays still between their researches and applications. Herein, we summarized the recent development in these three major fields which are indispensable in liver tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Hepatocytes/cytology , Liver/cytology , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bioreactors , Humans , Liver/growth & development , Liver, Artificial
15.
Brain Res ; 1634: 140-149, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26794251

ABSTRACT

Although studies have shown that cerebral ischemic preconditioning (IPC) can ameliorate ischemia/reperfusion (I/R) induced brain damage, but its precise mechanisms remain unknown. Therefore, the aim of this study was to investigate the neuroprotective mechanisms of IPC against ischemic brain damage induced by cerebral I/R and to explore whether the Calcium/calmodulin-dependent protein kinase II (CaMKII)-mediated up-regulation of nNOS ser847-phosphorylation signaling pathway contributed to the protection provided by IPC. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. The rats were pretreated with 3 min of IPC alone or KN62 (selective antagonist of CaMKII) treatment before IPC, after reperfusion for 3 days, 6 min ischemia was induced. Cresyl violet staining was used to examine the survival of hippocampal CA1 pyramidal neurons. Immunoblotting was performed to measure the phosphorylation of CaMKII, nNOS, c-Jun and the expression of FasL. Immunoprecipitation was used to examine the binding between PSD95 and nNOS. The results showed that IPC could significantly protect neurons against cerebral I/R injury, furthermore, the combination of PSD95 and nNOS was increased, coinstantaneously the phosphorylation of CaMKII and nNOS (ser847) were up-regulated, however the activation of c-Jun and FasL were reduced. Conversely, KN62 treatment before IPC reversed all these effects of IPC. Taken together, the results suggest that IPC could diminish ischemic brain injury through CaMKII-mediated up-regulation of nNOS ser847-phosphorylation signaling pathway.


Subject(s)
Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Ischemic Preconditioning , Nitric Oxide Synthase Type I/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Apoptosis/drug effects , CA1 Region, Hippocampal/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Disks Large Homolog 4 Protein , Fas Ligand Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Membrane Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Signal Transduction/drug effects
16.
Cell Prolif ; 48(2): 157-65, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25656979

ABSTRACT

End-stage liver disease can be the termination of acute or chronic liver diseases, with manifestations of liver failure; transplantation is currently an effective treatment for these. However, transplantation is severely limited due to the serious lack of donors, expense, graft rejection and requirement of long-term immunosuppression. Mesenchymal stem cells (MSCs) have attracted considerable attention as therapeutic tools as they can be obtained with relative ease and expanded in culture, along with features of self-renewal and multidirectional differentiation. Many scientific groups have sought to use MSCs differentiating into functional hepatocytes to be used in cell transplantation with liver tissue engineering to repair diseased organs. In most of the literature, hepatocyte differentiation refers to use of various additional growth factors and cytokines, such as hepatocyte growth factor (HGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), oncostatin M (OSM) and more, and most are involved in signalling pathway regulation and cell-cell/cell-matrix interactions. Signalling pathways have been shown to play critical roles in embryonic development, tumourigenesis, tumour progression, apoptosis and cell-fate determination. However, mechanisms of MSCs differentiating into hepatocytes, particularly signalling pathways involved, have not as yet been completely illustrated. In this review, we have focused on progress of signalling pathways associated with mesenchymal stem cells differentiating into hepatocytes along with the stepwise differentiation procedure.


Subject(s)
Cell Differentiation/physiology , Cell- and Tissue-Based Therapy , Hepatocytes/cytology , Mesenchymal Stem Cells/cytology , Cell Proliferation , End Stage Liver Disease/therapy , Humans , Liver/metabolism , Mesenchymal Stem Cell Transplantation , Signal Transduction , Tissue Engineering
17.
Biomed Mater Eng ; 25(1 Suppl): 137-43, 2015.
Article in English | MEDLINE | ID: mdl-25538064

ABSTRACT

The shortage of organ resource has been limiting the application of liver transplantation. Bioartificial liver construction is increasingly focused as a replacement treatment. To product a bioartificial liver, three elements must be considered: seeding cells, scaffold and bioreactor. Recent studies have shown that several methods can successfully differentiate MSC (mesenchymal stem cells) derived from Wharton's jelly into hepatocyte, such as stimulating MSC by cytokines and growth factors, direct and indirect co-culture MSC with hepatocytes, or promote MSC differentiation by 3-dimensional matrix. In some cases, differentiation of MSC into hepatocytes can also be an alternative approach for whole organ transplantation in treatment of acute and chronic liver diseases. In this review, the characterization of MSC from Wharton's jelly, their potential of application in liver tissue engineering on base of decellularized scaffold, their status of banking and their preclinical work performed will be discussed.


Subject(s)
Liver, Artificial , Mesenchymal Stem Cells/cytology , Organ Culture Techniques/instrumentation , Tissue Engineering/instrumentation , Tissue Scaffolds , Wharton Jelly/cytology , Bioreactors , Cell Differentiation/physiology , Cells, Cultured , Humans , Mesenchymal Stem Cell Transplantation/instrumentation , Mesenchymal Stem Cells/physiology , Prosthesis Design
18.
Biomed Mater Eng ; 25(1 Suppl): 159-66, 2015.
Article in English | MEDLINE | ID: mdl-25538066

ABSTRACT

OBJECTIVES: In present study, we plan to produce a decellularization protocol from rat liver to generate a three-dimensional whole organ scaffold. METHODS: A combination of 1% SDS and 1% tritonX-100 were used orderly to decellularize rat livers. After about 6 h of interactive antegrade/retrograde perfusion, a decellularized whole translucent liver scaffold with integrated blood vessel networks was generated. The decellularized livers are charactered by light microscopy, scanning electron microscopy, and biochemical analysis (DNA quantification) for preservation of the three-dimension of extracellular matrix architecture. RESULTS: The decellularization protocol was verified by observation of the whole translucent liver organ with intact vascular trees under macroscopy, in conjunction with the hematoxylin-eosin staining that showed no cells or nuclear material remained. Additionally, the Masson's stain indicted that the extracellular proteins were well kept and scanning electron microscopy (SEM) revealed a preserved decellularized matrix architecture. Compared to normal livers, DNA in the decellularized livers was quantified less than 10% at the same mass. CONCLUSIONS: The current method of decellularization protocol was feasible, simple and quick, and was verified by an absence of residual cells. The decellularized extracellular matrix had preserved integrate vascular network and a three-dimensional architecture.


Subject(s)
Extracellular Matrix/chemistry , Liver, Artificial , Liver/chemistry , Liver/ultrastructure , Tissue Engineering/instrumentation , Tissue Scaffolds , Animals , Cell-Free System , Equipment Design , Equipment Failure Analysis , Extracellular Matrix/ultrastructure , Female , Rats , Rats, Sprague-Dawley
19.
Biomed Mater Eng ; 25(1 Suppl): 167-77, 2015.
Article in English | MEDLINE | ID: mdl-25538067

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs) and myeloid-derived suppressor cells (MDSCs) can be mobilized from bone marrow (BM) into blood stream and home in tumor stroma, where they either help or hinder tumor growth. The issue of whether BMSCs could affect MDSCs in ascitogenous hepatoma BALB/c mice, thus influencing their functional activity, remains unclear. In this study, we demonstrated that after transfusion into ascitogenous hepatoma BALB/c mice, the homing fraction of BMSCs in BM was 2%-5% in 24-72 h and the percentage of Gr-1+CD11b+ MDSCs was downregulated in peripheral blood (PB) and BM. Meanwhile, IFN-γ+ T lymphocytes in PB increased. As a result of such immunoregulation, BMSCs treatment caused a delayed tumor growth and a prolonged survival in H22 ascitogenous hepatoma model. Because the proliferation of H22 cells was not affected by in vitro coculture with BMSCs, this observation is likely due to a systemic suppressive effect on the host MDSCs. We also demonstrated that BMSCs inhibited the induction and proliferation of MDSCs from hematopoietic stem cells (HSCs) in an in vitro tumor conditioned medium. Thus, our findings show for the first time that BMSCs are potentially inhibitor during MDSCs induction and proliferation and that when injected intravenously into tumor bearing mice they might be effective antitumor agents suitable for cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mesenchymal Stem Cell Transplantation/methods , T-Lymphocytes, Regulatory/pathology , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Carcinoma, Hepatocellular/immunology , Cell Communication/immunology , Cell Proliferation , Disease Progression , Liver Neoplasms/immunology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/immunology , Treatment Outcome
20.
Biomed Mater Eng ; 22(1-3): 105-11, 2012.
Article in English | MEDLINE | ID: mdl-22766708

ABSTRACT

Liver diseases have become one of the most important causes of morbidity and mortality in the world. Cell therapy and liver transplantation are though to be two treatment options well accepted. However, the shortage of cells sources in cytotherapy and the lack of liver donor in liver transplantation are the major obstacles for the performance of these treatment methods. It urged us to find new origins of extra-hepatic cells. A number of recent studies show that extra-hepatic mesenchymal stem cells (MSC) from different tissues can be differentiated into hepatocytes like cells (HLC). Several hepatic differentiation protocols of MSC have been published in recent years, based on cellular stimulation with exogenous cytokines/growth factors, co-culture with fetal or adult hepatocytes, 2- or 3-dimensional (2D, 3D) matrices to favor differentiation. Independently from the starting stem cells population used, some minimal criteria must be fulfilled to ensure therapeutic success: in vitro expandability, expression of hepatic like surface markers, with hepatic cell functions, and minimal or absent immunogenicity in the recipient host. In this review, we focused on stem cells originated from bone marrow, umbilical cord and adipose tissue which are widely investigated in recent years and have been proved to have liver regenerative potential, the factors used to differentiate stem cells to hepatocyte-like cells and the methods used to investigate these cells.


Subject(s)
Hepatocytes/cytology , Liver Regeneration , Liver/physiology , Mesenchymal Stem Cells/cytology , Adipose Tissue/cytology , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Humans , Liver/cytology , Umbilical Cord/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...