Zeitschriftenartikel zum Thema „Hamsoch“

Um die anderen Arten von Veröffentlichungen zu diesem Thema anzuzeigen, folgen Sie diesem Link: Hamsoch.

Geben Sie eine Quelle nach APA, MLA, Chicago, Harvard und anderen Zitierweisen an

Wählen Sie eine Art der Quelle aus:

Machen Sie sich mit Top-50 Zeitschriftenartikel für die Forschung zum Thema "Hamsoch" bekannt.

Neben jedem Werk im Literaturverzeichnis ist die Option "Zur Bibliographie hinzufügen" verfügbar. Nutzen Sie sie, wird Ihre bibliographische Angabe des gewählten Werkes nach der nötigen Zitierweise (APA, MLA, Harvard, Chicago, Vancouver usw.) automatisch gestaltet.

Sie können auch den vollen Text der wissenschaftlichen Publikation im PDF-Format herunterladen und eine Online-Annotation der Arbeit lesen, wenn die relevanten Parameter in den Metadaten verfügbar sind.

Sehen Sie die Zeitschriftenartikel für verschiedene Spezialgebieten durch und erstellen Sie Ihre Bibliographie auf korrekte Weise.

1

You, Qi, Ziming Liu, Jun Zhang, Mengjie Shen, Yuwan Li, Ying Jin und Yi Liu. „Human Amniotic Mesenchymal Stem Cell Sheets Encapsulating Cartilage Particles Facilitate Repair of Rabbit Osteochondral Defects“. American Journal of Sports Medicine 48, Nr. 3 (15.01.2020): 599–611. http://dx.doi.org/10.1177/0363546519897912.

Der volle Inhalt der Quelle
Annotation:
Background: Human amniotic mesenchymal stem cells (hAMSCs) are being widely applied in various fields. Therefore, hAMSCs represent a promising candidate to facilitate cartilage regeneration. Nonetheless, no studies have investigated the application of hAMSC sheets to repair cartilage defects in vivo. Purpose: To evaluate hAMSC sheets encapsulating cartilage particles to promote repair of rabbit osteochondral defects. Study Design: Controlled laboratory study. Methods: hAMSC sheets were constructed with passage 3 hAMSCs. The phenotypic and structural characteristics of hAMSC sheets were evaluated by flow cytometry and scanning electron microscopy, respectively. The potential for chondrogenic differentiation of hAMSC sheets was assessed by cartilage-specific marker staining, immunohistochemistry, and mRNA and protein expression (SOX9, COLII, and ACAN). Osteochondral defects (diameter, 3.5 mm; depth, 3 mm) were created in the left patellar grooves of 20 New Zealand White rabbits (female or male). The defects were treated with hAMSC sheet/cartilage particles (n = 5), cartilage particles (n = 5), hAMSC sheets (n = 5), or fibrin glue (n = 5). Macroscopic and histological evaluations of the regenerated tissue were conducted after 3 months. The survival time and differentiation of transplanted hAMSCs in the defect area were evaluated by immunofluorescence. Results: hAMSC sheets had a multilayered structure, with cells stacked layer by layer. Importantly, hAMSC sheets highly expressed phenotypic markers of mesenchymal stem cells. Cartilage-specific marker staining and immunohistochemistry were positive, and mRNA and protein expression was higher in the chondrogenically induced hAMSC sheet group than in the hAMSC sheet group ( P < .05). hAMSC sheet/cartilage particles formed a large amount of hyaline-like cartilage in the defect area. In addition, macroscopic and histological scores were significantly higher than those in the other groups. Integration with surrounding normal cartilage and subchondral bone regeneration in the hAMSC sheet/cartilage particles group were better when compared with the other groups. A large number of human nuclear-specific antigen-positive cells were observed in the defect area of hAMSC sheet/cartilage particles and hAMSC sheet groups. Moreover, some positive cells expressed SOX9. Conclusion: hAMSC sheets encapsulating cartilage particles facilitate osteochondral defect repair. Clinical Relevance: Delivery of cells in the form of a cell sheet in conjunction with cartilage particles provides a novel approach for cell-based cartilage regeneration.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
2

Hur, Junseok W., Min-Sik Kim, Se-Yeon Oh, Ho-Young Kang, Jingi Bae, Hokeun Kim, Hangyeore Lee, Sang-Won Lee und Dong-Hyuk Park. „Label-Free Quantitative Proteome Profiling of Cerebrospinal Fluid from a Rat Stroke Model with Stem Cell Therapy“. Cell Transplantation 30 (01.01.2021): 096368972110234. http://dx.doi.org/10.1177/09636897211023474.

Der volle Inhalt der Quelle
Annotation:
Human adipose-derived mesenchymal stem cells (hAMSCs) are capable of immunomodulation and regeneration after neural injury. For these reasons, hAMSCs have been investigated as a promising stem cell candidate for stroke treatment. However, noninvasive experiments studying the effects of grafted stem cells in the host brain have not yet been reported. Cerebrospinal fluid (CSF), which can be collected without sacrificing the subject, is involved in physiological control of the brain and reflects the pathophysiology of various neurological disorders of the central nervous system (CNS). Following stem cell transplantation in a stroke model, quantitative analysis of CSF proteome changes can potentially reveal the therapeutic effect of stem cells on the host CNS. We examined hAMSC-secreted proteins obtained from serum-free culture medium by liquid chromatography-tandem mass spectrometry (LC-MS/MS), which identified several extracellular matrix proteins, supporting the well-known active paracrine function of hAMSCs. Subsequently, we performed label-free quantitative proteomic analysis on CSF samples from rat stroke models intravenously injected with hAMSC (experimental) or phosphate buffered saline (control). In total, 524 proteins were identified; among them, 125 and 91 proteins were increased and decreased with hAMSC treatment, respectively. Furthermore, gene set enrichment analysis revealed three proteins, 14-3-3 theta, MAG, and neurocan, that showed significant increases in the hAMSC-treated model; these proteins are core members of neurotrophin signaling, nerve growth factor (NGF) signaling, and glycosaminoglycan metabolism, respectively. Subsequent histological and neurologic function experiments validated proliferative neurogenesis in the hAMSC-treated stroke model. We conclude that (i) intravenous injection of hAMSCs can induce neurologic recovery in a rat stroke model and (ii) CSF may reflect the therapeutic effect of hAMSCs. Additionally, proteins as 14-3-3 theta, MAG, and neurocan could be considered as potential CSF biomarkers of neuroregeneration. These CSF proteome profiling results would be utilized as valuable resource in further stroke studies.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
3

Yin, Lu, Zhi-xuan Zhou, Ming Shen, Ning Chen, Fei Jiang und Shou-Lin Wang. „The Human Amniotic Mesenchymal Stem Cells (hAMSCs) Improve the Implant Osseointegration and Bone Regeneration in Maxillary Sinus Floor Elevation in Rabbits“. Stem Cells International 2019 (11.12.2019): 1–10. http://dx.doi.org/10.1155/2019/9845497.

Der volle Inhalt der Quelle
Annotation:
Insufficient bone height in the posterior maxilla is a challenging problem in dental implantation. Bio-Oss, though routinely used in maxillary sinus floor elevation (MSFE), is not osteoinductive. Human amniotic mesenchymal cells (hAMSCs) isolated from placental tissues have potential for multidifferentiation and immunomodulatory properties and can be easily obtained without the need for invasive procedures and without ethical concerns. This is the first study to use hAMSCs to improve implant osseointegration and bone regeneration after MSFE. Human AMSCs were loaded into a fibrin gel and injected into rabbit MSFE models. The rabbits were assigned to four groups (n=3 per group), i.e., the control group, the hAMSC group, the Bio-Oss group, and the hAMSC/Bio-Oss group. The animals were sacrificed at postsurgery for four and twelve weeks and evaluated by histology and immunohistochemistry. Bone volume, bone volume/tissue volume, bone-to-implant contact ratio, and vessel-like structures in the hAMSC/Bio-Oss group were significantly better than those in other groups in the peri-implant and augmented areas. Immunofluorescence staining showed that alkaline phosphatase (ALP) activities of two hAMSC groups were higher than those of the other two groups. Sequential fluorescent labeling was performed in all of the 12-week groups. Observations showed that hAMSCs accelerated mineralized deposition rates on implant surfaces and in bone-augmented areas. These data demonstrated that hAMSCs could enhance implant osseointegration and bone regeneration after MSFE and might be used to optimize dental implantation in the future.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
4

Miceli, Vitale, Alessandro Bertani, Cinzia Maria Chinnici, Matteo Bulati, Mariangela Pampalone, Giandomenico Amico, Claudia Carcione, Eva Schmelzer, Jörg C. Gerlach und Pier Giulio Conaldi. „Conditioned Medium from Human Amnion-Derived Mesenchymal Stromal/Stem Cells Attenuating the Effects of Cold Ischemia-Reperfusion Injury in an In Vitro Model Using Human Alveolar Epithelial Cells“. International Journal of Molecular Sciences 22, Nr. 2 (06.01.2021): 510. http://dx.doi.org/10.3390/ijms22020510.

Der volle Inhalt der Quelle
Annotation:
The clinical results of lung transplantation (LTx) are still less favorable than other solid organ transplants in both the early and long term. The fragility of the lungs limits the procurement rate and can favor the occurrence of ischemia-reperfusion injury (IRI). Ex vivo lung perfusion (EVLP) with Steen SolutionTM (SS) aims to address problems, and the implementation of EVLP to alleviate the activation of IRI-mediated processes has been achieved using mesenchymal stromal/stem cell (MSC)-based treatments. In this study, we investigated the paracrine effects of human amnion-derived MSCs (hAMSCs) in an in vitro model of lung IRI that includes cold ischemia and normothermic EVLP. We found that SS enriched by a hAMSC-conditioned medium (hAMSC-CM) preserved the viability and delayed the apoptosis of alveolar epithelial cells (A549) through the downregulation of inflammatory factors and the upregulation of antiapoptotic factors. These effects were more evident using the CM of 3D hAMSC cultures, which contained an increased amount of immunosuppressive and growth factors compared to both 2D cultures and encapsulated-hAMSCs. To conclude, we demonstrated an in vitro model of lung IRI and provided evidence that a hAMSC-CM attenuated IRI effects by improving the efficacy of EVLP, leading to strategies for a potential implementation of this technique.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
5

Jiang, Fei, Jie Ma, Yi Liang, Yuming Niu, Ning Chen und Ming Shen. „Amniotic Mesenchymal Stem Cells Can Enhance Angiogenic Capacity via MMPsIn VitroandIn Vivo“. BioMed Research International 2015 (2015): 1–15. http://dx.doi.org/10.1155/2015/324014.

Der volle Inhalt der Quelle
Annotation:
The aim of this study was to evaluate the angiogenic capacity and proteolytic mechanism of coculture using human amniotic mesenchymal stem cells (hAMSCs) with human umbilical vein endothelial cells (HUVECs)in vivoandin vitroby comparing to those of coculture using bone marrow mesenchymal stem cells with HUVEC. For thein vivoexperiment, cells (HUVEC-monoculture, HUVEC-hAMSC coculture, and HUVEC-BMMSC coculture) were seeded in fibrin gels and injected subcutaneously in nude mice. The samples were collected on days 7 and 14 and histologically analyzed by H&E and CD31 staining. CD31-positive staining percentage and vessel-like structure (VLS) density were evaluated as quantitative parameters for angiogenesis. The increases of CD31-positive staining area and VLS density in both HUVEC-hAMSC group and HUVEC-BMMSC group were found between two time points, while obvious decline of those was observed in HUVEC-only group. For thein vitroexperiment, we utilized the same 3D culture model to investigate the proteolytic mechanism related to capillary formation. Intensive vascular networks formed by HUVECs were associated with hAMSCs or BMMSCs and related to MMP2 and MMP9. In conclusion, hAMSCs shared similar capacity and proteolytic mechanism with BMMSCs on neovascularization.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
6

Sandonà, Martina, Federica Esposito, Anna Cargnoni, Antonietta Silini, Pietro Romele, Ornella Parolini und Valentina Saccone. „Amniotic Membrane-Derived Stromal Cells Release Extracellular Vesicles That Favor Regeneration of Dystrophic Skeletal Muscles“. International Journal of Molecular Sciences 24, Nr. 15 (05.08.2023): 12457. http://dx.doi.org/10.3390/ijms241512457.

Der volle Inhalt der Quelle
Annotation:
Duchenne muscular dystrophy (DMD) is a muscle disease caused by mutations in the dystrophin gene characterized by myofiber fragility and progressive muscle degeneration. The genetic defect results in a reduced number of self-renewing muscle stem cells (MuSCs) and an impairment of their activation and differentiation, which lead to the exhaustion of skeletal muscle regeneration potential and muscle replacement by fibrotic and fatty tissue. In this study, we focused on an unexplored strategy to improve MuSC function and to preserve their niche based on the regenerative properties of mesenchymal stromal cells from the amniotic membrane (hAMSCs), that are multipotent cells recognized to have a role in tissue repair in different disease models. We demonstrate that the hAMSC secretome (CM hAMSC) and extracellular vesicles (EVs) isolated thereof directly stimulate the in vitro proliferation and differentiation of human myoblasts and mouse MuSC from dystrophic muscles. Furthermore, we demonstrate that hAMSC secreted factors modulate the muscle stem cell niche in dystrophic–mdx-mice. Interestingly, local injection of EV hAMSC in mdx muscles correlated with an increase in the number of activated Pax7+/Ki67+ MuSCs and in new fiber formation. EV hAMSCs also significantly reduced muscle collagen deposition, thus counteracting fibrosis and MuSCs exhaustion, two hallmarks of DMD. Herein for the first time we demonstrate that CM hAMSC and EVs derived thereof promote muscle regeneration by supporting proliferation and differentiation of resident muscle stem cells. These results pave the way for the development of a novel treatment to counteract DMD progression by reducing fibrosis and enhancing myogenesis in dystrophic muscles.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
7

Sun, Shoujia, Quan Zhang, Man Li, Pan Gao, Kuan Huang, Rajluxmee Beejadhursing, Wei Jiang, Ting Lei, Mingxin Zhu und Kai Shu. „GDNF Promotes Survival and Therapeutic Efficacy of Human Adipose-Derived Mesenchymal Stem Cells in a Mouse Model of Parkinson’s Disease“. Cell Transplantation 29 (01.01.2020): 096368972090851. http://dx.doi.org/10.1177/0963689720908512.

Der volle Inhalt der Quelle
Annotation:
Mesenchymal stem cell (MSC)-based regenerative therapy is regarded as a promising strategy for the treatment of Parkinson’s disease (PD). However, MSC components may exhibit poor intracranial survivability, particularly in the later stages following cell transplantation, limiting their potential curative effect and also clinical applications. Glial cell line-derived neurotrophic factor (GDNF), which encompasses a variety of transforming growth factor beta super family members, has been reported to enhance motor function and exert neuroprotective effects. However, no previous studies have investigated the effects of GDNF on human primary adipose-derived MSCs (hAMSCs), despite its potential for enhancing stem cell survival and promoting therapeutic efficacy in the treatment of PD. In the present study, we proposed a novel approach for enhancing the proliferative capacity and improving the efficacy of hAMSC treatment. Pre-exposure of engineered hAMSCs to GDNF enhanced the proliferation and differentiation of these stem cells in vitro. In addition, in 6-hydroxydopamine-lesioned mice, a common PD model, intracranial injection of hAMSCs-GDNF was associated with greater performance on behavioral tests, larger graft volumes 5 weeks after transplantation, and higher levels of Nestin, glial fibrillary acidic protein, and Tuj-1 differentiation than those treated with hAMSCs-Vector. Following transplantation of hAMSCs-GDNF into the striatum of lesioned models, we observed significant increases in tyrosine hydroxylase- and NeuN-positive staining. These findings highlight the therapeutic potential of hAMSCs-GDNF for patients with PD, as well as an efficient method for promoting therapeutic efficacy of these delivery vehicles.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
8

Fu, Qingjie, Shunsuke Ohnishi und Naoya Sakamoto. „Conditioned Medium from Human Amnion-Derived Mesenchymal Stem Cells Regulates Activation of Primary Hepatic Stellate Cells“. Stem Cells International 2018 (08.10.2018): 1–11. http://dx.doi.org/10.1155/2018/4898152.

Der volle Inhalt der Quelle
Annotation:
Mesenchymal stem cells (MSCs), or multipotent mesenchymal stromal cells, are present in almost all organs and tissues, including the amnion. Human amnion-derived mesenchymal stem cell (hAMSC) transplantation has been reported to ameliorate liver fibrosis in animal models. However, the mechanism for the prevention of liver fibrosis is poorly understood. In this study, we investigated the effects, and underlying mechanisms, of a conditioned medium obtained from hAMSC cultures (hAMSC-CM) on a primary culture of rat hepatic stellate cells (HSCs). We observed that in routine culture, hAMSC-CM in HSCs significantly inhibited the expression of alpha-smooth muscle actin (α-SMA), an activation marker of HSCs, and the production of collagen type 1 (COL1), a dominant component of the extracellular matrix (ECM) in the culture medium. In addition, hAMSC-CM upregulated the expression of ECM degradation-related genes, such as metalloproteinase- (Mmp-) 2, Mmp-9, Mmp-13, and tissue inhibitor of metalloproteinase- (Timp-) 1; however, it did not affect the expression of collagen type 1α1 (Col1a1). These regulatory effects on HSCs were concentration-dependent. A cell proliferation assay indicated that hAMSC-CM significantly suppressed HSC proliferation and downregulated the expression of cyclin B (Ccnb), a proliferation-related gene. Transforming growth factor-beta (TGF-β) treatment further activated HSCs and hAMSC-CM significantly inhibited the upregulation of α-Sma and Col1a1 induced by TGF-β. These findings demonstrated that hAMSC-CM can modulate HSC function via secretory factors and provide a plausible explanation for the protective role of hAMSCs in liver fibrosis.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
9

Fioretti, Daniela, Mario Ledda, Sandra Iurescia, Raffaella Carletti, Cira Di Gioia, Maria Grazia Lolli, Rodolfo Marchese, Antonella Lisi und Monica Rinaldi. „Severely Damaged Freeze-Injured Skeletal Muscle Reveals Functional Impairment, Inadequate Repair, and Opportunity for Human Stem Cell Application“. Biomedicines 12, Nr. 1 (21.12.2023): 30. http://dx.doi.org/10.3390/biomedicines12010030.

Der volle Inhalt der Quelle
Annotation:
Background: The regeneration of severe traumatic muscle injuries is an unsolved medical need that is relevant for civilian and military medicine. In this work, we produced a critically sized nonhealing muscle defect in a mouse model to investigate muscle degeneration/healing phases. Materials and methods: We caused a freeze injury (FI) in the biceps femoris of C57BL/6N mice. From day 1 to day 25 post-injury, we conducted histological/morphometric examinations, an analysis of the expression of genes involved in inflammation/regeneration, and an in vivo functional evaluation. Results: We found that FI activates cytosolic DNA sensing and inflammatory responses. Persistent macrophage infiltration, the prolonged expression of eMHC, the presence of centrally nucleated myofibers, and the presence of PAX7+ satellite cells at late time points and with chronic physical impairment indicated inadequate repair. By looking at stem-cell-based therapeutic protocols of muscle repair, we investigated the crosstalk between M1-biased macrophages and human amniotic mesenchymal stem cells (hAMSCs) in vitro. We demonstrated their reciprocal paracrine effects where hAMSCs induced a shift of M1 macrophages into an anti-inflammatory phenotype, and M1 macrophages promoted an increase in the expression of hAMSC immunomodulatory factors. Conclusions: Our findings support the rationale for the future use of our injury model to exploit the full potential of in vivo hAMSC transplantation following severe traumatic injuries.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
10

Kamadjaja, David. „The Osteogenic Capacity of HumanAmniotic Membrane Mesenchymal Stem Cell (hAMSC) and Potential for Application in Maxillofacial Bone Reconstruction in Vitro Study“. Journal of Stem Cell Research and Tissue Engineering 4, Nr. 1 (26.08.2020): 17. http://dx.doi.org/10.20473/jscrte.v4i1.21590.

Der volle Inhalt der Quelle
Annotation:
Amniotic membrane of human placenta is a source of abundant mesenchymal stem cell (hAMSC) which makes it a potential source of allogeneic multipotent cell for bone healing. However, much has to be explored about its isolation procedure and the osteogenic differentiation potential. The aims of this study are to establish the procurement procedure of human amniotic membrane, the isolation and culture of hAMSC, the MSC phenotypic characterization, and the in vitro osteogenic differentiation of hAMSC. Results of the study are as follows. The quality of human amniotic membrane would be best if procured from Caesarean operation under highly aseptic condition to avoid fungal and bacterial contamination on the culture. Isolation procedure using modified Soncini protocol yielded large amount of MSC with high proliferative capacity in culture medium. Characterization of hAMSC showed that the majority of the target cells exhibited specific MSC markers (CD10S and CD90) with a small number of these cells expressing CD45the marker of hematopoeitic cells. The in vitro osteogenic differentiation of hAMSC followed by Alizarin Red staining showed that osteoblastic differentiation was detected in a significantly high number of cells. This study concludes that hAMSCs isolated from human amniotic membrane have the capacity for in vitro osteogenesis which makes them be one of the potential allogeneic stem cells for application in maxillofacial bone reconstruction.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
11

Xu, Ying, Ya Gao, Yan chun Yang, Dongmao Zhu, Yintian Zhang, Ke Zhao, Yu Zhang, Qifa Liu, Haitao Sun und Baohong Ping. „Immunoregulatory Properties of Apoptotic Human Amniontic Mesenchymal Stromal Cells“. Blood 132, Supplement 1 (29.11.2018): 5678. http://dx.doi.org/10.1182/blood-2018-99-112585.

Der volle Inhalt der Quelle
Annotation:
Abstract Objective Mesenchymal stromal cells (MSCs) have been used in preventing and treating acute graft-versus-host disease (aGVHD), but the mechanism is not fully understood. Apoptotic bone marrow mesenchymal stromal cells (BMSCs) were showed could induce vivo recipient-mediated immunomodulation in mice GVHD model. We had demonstrated that, similar to BM-MSCs, human amniontic mesenchymal stromal cells (hAMSCs) exhibit potent immunosuppressive and anti-inflammatory activities but possess a higher proliferation activity and clearer stem cell properties in vitro. This study focuses on the immunoregulatory properties of apoptotic human amniontic mesenchymal stromal cells (apo-hAMSCs) in an inflammatory microenvironment. Methods hAMSCs from human amniotic membrane were cultured with tissue mass cell culture. The cell phenotype of the 3rd passage were detected by flow cytometry. Transwell co-culture experiments and cell-cell contact co-culture experiments were conducted, consisting of hAMSCs and peripheral blood mononuclear cells stimulated with phytohemagglutinin (PHA-PBMCs), as the positive control group. While other groups were PBMCs without PHA and hAMSCs(PBMCs+hAMSCs), PBMCs and PHA (PHA-PBMCs), hAMSCs and PBMCs. For apoptosis evalution, the morphological features of hAMSCs were recorded in different groups, and apo-hAMSCs were analyzed by flow cytometry at 24 hours. The production of Interferon-γ (IFN-γ), transforming growth factor-β1 (TGF-β1), prostaglandin E2 (PGE-2), soluble human leukocyte antigen G (sHLA-G), Tumor necrosis factor-α(TNF-α) and interleukin-17A (IL-17A) in the co-culture supernatant was detected using enzyme-linked immunosorbent assay (ELISA), and kynurenine were dectected by spectrophotometer. CD4+CD25+FOXP3+ regulatory T cells (Tregs) in PBMC were analyaed by flow cytometry. Result hAMSCs expressed CD105, CD73, CD90, while not CD19, CD34, CD45, CD11b, HLA-DR. In the group of hAMSCs and PHA-PBMCs, the number of hAMSCs reduced. The morphological features were that cells shrinked, turned round, separated from the bottle and suspended in supernatant. However, hAMSCs in the groups of hAMSCs+PBMCs and hAMSCs stayed the same. Apoptosis in hAMSCs cultivated with PHA-PBMCs via transwell or cell-cell contact experiment increased compared with the group of hAMSCs+PBMCs (P<0.05) and hAMSCs (P<0.05). In the two co-culture experments, the secretion level of PGE-2, TGF-β1, sHLA-G, and KYN significantly increased in hAMSCs with PHA-PBMCs compared with hAMSC (P<0.05) and hAMSCs with PBMCs (P<0.05). The level of IFN-γ and TNF-α decreased in hAMSCs with PHA-PBMCs compared with PBMCs with PHA (P<0.05). While the level of IL-17A was significantly increase in hAMSCs with PHA-PBMCs compared with hAMSCs (P<0.05), hAMSCs with PBMCs (P<0.05) and PHA-PBMCs (P>0.05). Evident difference of CD4+CD25+FOXP3+ Tregs was shown between hAMSCs with PHA-PBMCs and PHA-PBMCs (P<0.05). Conclusion Activated, but not resting, PBMCs induce extensive early apoptosis in hAMSCs. And apoptosis in hAMSCs need inflammatory microenvirenment. Apoptotic hAMSCs still have immunoregulatory effects in cytokines and immune cells. Funding This work was supported by Natural Science Foundation of China (81701243), Key Sci-Tech Research Projects of Guangdong Province (2014A02021102), Natural Science Foundation of Guangdong Province, China (2014A030310373), the Pearl River S&T Nova Program of Guangzhou (201710010047). Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
12

Khlusov, Igor, Kristina Yurova, Valeria Shupletsova, Olga Khaziakhmatova, Vladimir Malashchenko, Valeriya Kudryavtseva, Marina Khlusova, Gleb Sukhorukov und Larisa Litvinova. „Microcapsule-Based Dose-Dependent Regulation of the Lifespan and Behavior of Adipose-Derived MSCs as a Cell-Mediated Delivery System: In Vitro Study“. International Journal of Molecular Sciences 24, Nr. 1 (24.12.2022): 292. http://dx.doi.org/10.3390/ijms24010292.

Der volle Inhalt der Quelle
Annotation:
The development of “biohybrid” drug delivery systems (DDS) based on mesenchymal stem/stromal cells (MSCs) is an important focus of current biotechnology research, particularly in the areas of oncotheranostics, regenerative medicine, and tissue bioengineering. However, the behavior of MSCs at sites of inflammation and tumor growth is relevant to potential tumor transformation, immunosuppression, the inhibition or stimulation of tumor growth, metastasis, and angiogenesis. Therefore, the concept was formulated to control the lifespan of MSCs for a specific time sufficient for drug delivery to the target tissue by varying the number of internalized microcontainers. The current study addressed the time-dependent in vitro assessment of the viability, migration, and division of human adipose-derived MSCs (hAMSCs) as a function of the dose of internalized polyelectrolyte microcapsules prepared using a layer-by-layer technique. Polystyrene sulfonate (PSS)—poly(allylamine hydrochloride) (PAH)-coated spherical micrometer-sized (diameter ~2–3 µm) vaterite (CaCO3) microcapsules (PAH-PSS)6 with the capping PSS layer were prepared after dissolution of the CaCO3 core template. The Cell-IQ phase contrast imaging results showed that hAMSCs internalized all (PAH-PSS)6 microcapsules saturating the intercellular medium (5–90 particles per cell). A strong (r > 0.7) linear dose-dependent and time-dependent (up to 8 days) regression was observed between the in vitro decrease in cell viability and the number of internalized microvesicles. The approximate time-to-complete-death of hAMSCs at different concentrations of microcapsules in culture was 428 h (1:5 ratio), 339 h (1:10), 252 h (1:20), 247 h (1:45), and 170 h (1:90 ratio). By varying the number of microcontainers loaded into the cells (from 1:10 to 1:90), a dose-dependent exponential decrease in both the movement rate and division rate of hAMSCs was observed. A real-time cell analysis (RTCA) of the effect of (PAH-PSS)6 microcapsules (from 1:5 to 1:20) on hAMSCs also showed a dose- and time-dependent decrease in cell longevity after a 50h study at ratios of 1:10 and 1:20. The incorporation of microcapsules (1:5, 1:20, and 1:45) resulted in a dose-dependent increase in 24-48h secretion of GRO-α (CXCL1), MIF, and SDF-1α (CXCL12) chemokines in hAMSC culture. In turn, the normalization or inhibition of chemokine secretion occurred after 72 h, except for MIF levels below 5–20 microcapsules, which were internalized by MSCs. Thus, the proposed concept of controlling the lifespan of MSC-based DDS using a dose of internalized PAH-PSS microcapsules could be useful for biomedical applications. (PAH-PSS)6 microcapsule ratios of 1:5 and 1:10 have little effect on the lifespan of hAMSCs for a long time (up to 14–18 days), which can be recommended for regenerative therapy and tissue bioengineering associated with low oncological risk. The microcapsule ratios of 1:20 and 1:45 did not significantly restrict the migratory activity of hAMSCs-based DDS during the time interval required for tissue delivery (up to 4–5 days), followed by cell death after 10 days. Therefore, such doses of microcapsules can be used for hAMSC-based DDS in oncotheranostics.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
13

Gallo, Alessia, Nicola Cuscino, Flavia Contino, Matteo Bulati, Mariangela Pampalone, Giandomenico Amico, Giovanni Zito et al. „Changes in the Transcriptome Profiles of Human Amnion-Derived Mesenchymal Stromal/Stem Cells Induced by Three-Dimensional Culture: A Potential Priming Strategy to Improve Their Properties“. International Journal of Molecular Sciences 23, Nr. 2 (13.01.2022): 863. http://dx.doi.org/10.3390/ijms23020863.

Der volle Inhalt der Quelle
Annotation:
Mesenchymal stromal/stem cells (MSCs) are believed to function in vivo as a homeostatic tool that shows therapeutic properties for tissue repair/regeneration. Conventionally, these cells are expanded in two-dimensional (2D) cultures, and, in that case, MSCs undergo genotypic/phenotypic changes resulting in a loss of their therapeutic capabilities. Moreover, several clinical trials using MSCs have shown controversial results with moderate/insufficient therapeutic responses. Different priming methods were tested to improve MSC effects, and three-dimensional (3D) culturing techniques were also examined. MSC spheroids display increased therapeutic properties, and, in this context, it is crucial to understand molecular changes underlying spheroid generation. To address these limitations, we performed RNA-seq on human amnion-derived MSCs (hAMSCs) cultured in both 2D and 3D conditions and examined the transcriptome changes associated with hAMSC spheroid formation. We found a large number of 3D culture-sensitive genes and identified selected genes related to 3D hAMSC therapeutic effects. In particular, we observed that these genes can regulate proliferation/differentiation, as well as immunomodulatory and angiogenic processes. We validated RNA-seq results by qRT-PCR and methylome analysis and investigation of secreted factors. Overall, our results showed that hAMSC spheroid culture represents a promising approach to cell-based therapy that could significantly impact hAMSC application in the field of regenerative medicine.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
14

Ragni, Enrico, Carlotta Perucca Orfei, Antonietta Rosa Silini, Alessandra Colombini, Marco Viganò, Ornella Parolini und Laura de Girolamo. „miRNA Reference Genes in Extracellular Vesicles Released from Amniotic Membrane-Derived Mesenchymal Stromal Cells“. Pharmaceutics 12, Nr. 4 (11.04.2020): 347. http://dx.doi.org/10.3390/pharmaceutics12040347.

Der volle Inhalt der Quelle
Annotation:
Human amniotic membrane and amniotic membrane-derived mesenchymal stromal cells (hAMSCs) have produced promising results in regenerative medicine, especially for the treatment of inflammatory-based diseases and for different injuries including those in the orthopedic field such as tendon disorders. hAMSCs have been proposed to exert their anti-inflammatory and healing potential via secreted factors, both free and conveyed within extracellular vesicles (EVs). In particular, EV miRNAs are considered privileged players due to their impact on target cells and tissues, and their future use as therapeutic molecules is being intensely investigated. In this view, EV-miRNA quantification in either research or future clinical products has emerged as a crucial paradigm, although, to date, largely unsolved due to lack of reliable reference genes (RGs). In this study, a panel of thirteen putative miRNA RGs (let-7a-5p, miR-16-5p, miR-22-5p, miR-23a-3p, miR-26a-5p, miR-29a-5p, miR-101-3p, miR-103a-3p, miR-221-3p, miR-423-5p, miR-425-5p, miR-660-5p and U6 snRNA) that were identified in different EV types was assessed in hAMSC-EVs. A validated experimental pipeline was followed, sifting the output of four largely accepted algorithms for RG prediction (geNorm, NormFinder, BestKeeper and ΔCt method). Out of nine RGs constitutively expressed across all EV isolates, miR-101-3p and miR-22-5p resulted in the most stable RGs, whereas miR-423-5p and U6 snRNA performed poorly. miR-22-5p was also previously reported to be a reliable RG in adipose-derived MSC-EVs, suggesting its suitability across samples isolated from different MSC types. Further, to shed light on the impact of incorrect RG choice, the level of five tendon-related miRNAs (miR-29a-3p, miR-135a-5p, miR-146a-5p, miR-337-3p, let-7d-5p) was compared among hAMSC-EVs isolates. The use of miR-423-5p and U6 snRNA did not allow a correct quantification of miRNA incorporation in EVs, leading to less accurate fingerprinting and, if used for potency prediction, misleading indication of the most appropriate clinical batch. These results emphasize the crucial importance of RG choice for EV-miRNAs in hAMSCs studies and contribute to the identification of reliable RGs such as miR-101-3p and miR-22-5p to be validated in other MSC-EVs related fields.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
15

Xu, Yan, Huan Yuan, Yi Luo, Yu-Jie Zhao und Jian-Hui Xiao. „Ganoderic Acid D Protects Human Amniotic Mesenchymal Stem Cells against Oxidative Stress-Induced Senescence through the PERK/NRF2 Signaling Pathway“. Oxidative Medicine and Cellular Longevity 2020 (28.07.2020): 1–18. http://dx.doi.org/10.1155/2020/8291413.

Der volle Inhalt der Quelle
Annotation:
Aging is an important risk factor in the occurrence of many chronic diseases. Senescence and exhaustion of adult stem cells are considered as a hallmark of aging in organisms. In this study, a senescent human amniotic mesenchymal stem cell (hAMSC) model subjected to oxidative stress was established in vitro using hydrogen peroxide. We investigated the effects of ganoderic acid D (GA-D), a natural triterpenoid compound produced from Ganoderma lucidum, on hAMSC senescence. GA-D significantly inhibited β-galactosidase (a senescence-associated marker) formation, in a dose-dependent manner, with doses ranging from 0.1 μM to 10 μM, without inducing cytotoxic side-effects. Furthermore, GA-D markedly inhibited the generation of reactive oxygen species (ROS) and the expression of p21 and p16 proteins, relieved the cell cycle arrest, and enhanced telomerase activity in senescent hAMSCs. Furthermore, GA-D upregulated the expression of phosphorylated protein kinase R- (PKR-) like endoplasmic reticulum kinase (PERK), peroxidase III (PRDX3), and nuclear factor-erythroid 2-related factor (NRF2) and promoted intranuclear transfer of NRF2 in senescent cells. The PERK inhibitor GSK2656157 and/or the NRF2 inhibitor ML385 suppressed the PERK/NRF2 signaling, which was activated by GA-D. They induced a rebound for the generation of ROS and β-galactosidase-positive cells and attenuated the differentiation capacity. These findings suggest that GA-D retards hAMSC senescence through activation of the PERK/NRF2 signaling pathway and may be a promising candidate for the discovery of antiaging agents.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
16

Carbone, Annalucia, Roberto Zefferino, Elisa Beccia, Valeria Casavola, Stefano Castellani, Sante Di Gioia, Valentina Giannone et al. „Gap Junctions Are Involved in the Rescue of CFTR-Dependent Chloride Efflux by Amniotic Mesenchymal Stem Cells in Coculture with Cystic Fibrosis CFBE41o- Cells“. Stem Cells International 2018 (2018): 1–14. http://dx.doi.org/10.1155/2018/1203717.

Der volle Inhalt der Quelle
Annotation:
We previously found that human amniotic mesenchymal stem cells (hAMSCs) in coculture with CF immortalised airway epithelial cells (CFBE41o- line, CFBE) on Transwell® filters acquired an epithelial phenotype and led to the expression of a mature and functional CFTR protein. In order to explore the role of gap junction- (GJ-) mediated intercellular communication (GJIC) in this rescue, cocultures (hAMSC : CFBE, 1 : 5 ratio) were studied for the formation of GJIC, before and after silencing connexin 43 (Cx43), a major component of GJs. Functional GJs in cocultures were inhibited when the expression of the Cx43 protein was downregulated. Transfection of cocultures with siRNA against Cx43 resulted in the absence of specific CFTR signal on the apical membrane and reduction in the mature form of CFTR (band C), and in parallel, the CFTR-dependent chloride channel activity was significantly decreased. Cx43 downregulation determined also a decrease in transepithelial resistance and an increase in paracellular permeability as compared with control cocultures, implying that GJIC may regulate CFTR expression and function that in turn modulate airway epithelium tightness. These results indicate that GJIC is involved in the correction of CFTR chloride channel activity upon the acquisition of an epithelial phenotype by hAMSCs in coculture with CF cells.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
17

Suryawan, I. Gde Rurus, Anudya Kartika Ratri, Andrianto Andrianto, Meity Ardiana und Ricardo Adrian Nugraha. „Fibronectin enhances attachment of human adipose-derived mesenchymal stem cells into polytetrafluoroethylene patch during surgical closure of the atrial and ventricular septal defect“. Annals of Pediatric Cardiology 16, Nr. 3 (2023): 189–93. http://dx.doi.org/10.4103/apc.apc_9_23.

Der volle Inhalt der Quelle
Annotation:
ABSTRACT Background: Polytetrafluoroethylene (PTFE) patch is commonly used during surgical closure for atrial septal defect (ASD) and ventricular septal defect (VSD). However, this patch has several limitations such as its inability to grow or remodel, especially in children and young adults. To tackle these limitations, we have tried to use fibronectin and human adipose-derived mesenchymal stem cells (hAMSCs) in the PTFE patch. Objective: To understand the impact of fibronectin to enhance hAMSCs cell-to-cell adherence and cell-to-patch surface attachment into PTFE patches used in the surgical closure of ASD or VSD. Materials and Methods: The hAMSCs were plated and fixated with 15 mL methanol and cluster of differentiation (CD) 90+, CD105+, and CD45 − antibodies were labeled with fluorescein isothiocyanate, rinsed with phosphate-buffered saline, and analyzed under a fluorescence microscope. Fibronectin solution (0.1%) was used to soak patch scaffolds for approximately 2-h duration and then dried for 20 min in the treatment group. The samples were examined with a scanning electron microscope (SEM). Results: SEM examination showed incomplete attachment of the cells even after 10 days in the control group at 1.14 ± 1.13. In contrast, the treatment group showed more cells attached to the patch surface at 31.25 ± 13.28 (P ≤ 0.0001). The observation at 5 days was 17.67 ± 20.21, at 7 days was 12.11 ± 10.94, and at 10 days was 18.83 ± 23.25. There was no significant statistical difference in mean cell per view among each treatment group (P = 0.802). Conclusion: Our work demonstrates that fibronectin has a positive impact on hAMSC attachment seeded onto the PTFE patch. These properties, in combination with their developmental plasticity, have generated tremendous interest in regenerative medicine.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
18

Szychlinska, Marta Anna, Giovanna Calabrese, Silvia Ravalli, Nunziatina Laura Parrinello, Stefano Forte, Paola Castrogiovanni, Elisabetta Pricoco et al. „Cycloastragenol as an Exogenous Enhancer of Chondrogenic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells. A Morphological Study“. Cells 9, Nr. 2 (03.02.2020): 347. http://dx.doi.org/10.3390/cells9020347.

Der volle Inhalt der Quelle
Annotation:
Stem cell therapy and tissue engineering represent a promising approach for cartilage regeneration. However, they present limits in terms of mechanical properties and premature de-differentiation of engineered cartilage. Cycloastragenol (CAG), a triterpenoid saponin compound and a hydrolysis product of the main ingredient in Astragalus membranaceous, has been explored for cartilage regeneration. The aim of this study was to investigate CAG’s ability to promote cell proliferation, maintain cells in their stable active phenotype, and support the production of cartilaginous extracellular matrix (ECM) in human adipose-derived mesenchymal stem cells (hAMSCs) in up to 28 days of three-dimensional (3D) chondrogenic culture. The hAMSC pellets were cultured in chondrogenic medium (CM) and in CM supplemented with CAG (CAG–CM) for 7, 14, 21, and 28 days. At each time-point, the pellets were harvested for histological (hematoxylin and eosin (H&E)), histochemical (Alcian-Blue) and immunohistochemical analysis (Type I, II, and X collagen, aggrecan, SOX9, lubricin). After excluding CAG’s cytotoxicity (MTT Assay), improved cell condensation, higher glycosaminoglycans (sGAG) content, and increased cell proliferation have been detected in CAG–CM pellets until 28 days of culture. Overall, CAG improved the chondrogenic differentiation of hAMSCs, maintaining stable the active chondrocyte phenotype in up to 28 days of 3D in vitro chondrogenic culture. It is proposed that CAG might have a beneficial impact on cartilage regeneration approaches.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
19

Chen, Mei-ting, Yi-ting Zhao, Li-yuan Zhou, Ming Li, Qian Zhang, Qin Han und Xin-hua Xiao. „Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Enhance Insulin Sensitivity in Insulin Resistant Human Adipocytes“. Current Medical Science 41, Nr. 1 (Februar 2021): 87–93. http://dx.doi.org/10.1007/s11596-021-2323-4.

Der volle Inhalt der Quelle
Annotation:
SummaryInsulin resistance is an essential characteristic of type 2 diabetes mellitus (T2DM), which can be induced by glucotoxicity and adipose chronic inflammation. Mesenchymal stem cells (MSCs) and their exosomes were reported to ameliorate T2DM and its complications by their immunoregulatory and healing abilities. Exosomes derived from MSCs contain abundant molecules to mediate crosstalk between cells and mimic biological function of MSCs. But the role of exosomes derived from human umbilical cord mesenchymal stem cells (hUC-MSCs) in insulin resistance of human adipocytes is unclear. In this study, exosomes were harvested from the conditioned medium of hUC-MSCs and added to insulin-resistant adipocytes. Insulin-stimulated glucose uptake was measured by glucose oxidase/peroxidase assay. The signal pathway involved in exosome-treated adipocytes was detected by RT-PCR and Western blotting. The biological characteristics and function were compared between hUC-MSCs and human adipose-derived mesenchymal stem cells (hAMSCs). The results showed that hAMSCs had better adipogenic ability than hUC-MSCs. After induction of mature adipocytes by adipogenesis of hAMSC, the model of insulin-resistant adipocytes was successfully established by TNF-α and high glucose intervention. After exosome treatment, the insulin-stimulated glucose uptake was significantly increased. In addition, the effect of exosomes could be stabilized for at least 48 h. Furthermore, the level of leptin was significantly decreased, and the mRNA expression of sirtuin-1 and insulin receptor substrate-1 was significantly upregulated after exosome treatment. In conclusion, exosomes significantly improve insulin sensitivity in insulin-resistant human adipocytes, and the mechanism involves the regulation of adipokines.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
20

Feng, Y., M. Zhu, S. Dangelmajer, Y. M. Lee, O. Wijesekera, C. X. Castellanos, A. Denduluri et al. „Hypoxia-cultured human adipose-derived mesenchymal stem cells are non-oncogenic and have enhanced viability, motility, and tropism to brain cancer“. Cell Death & Disease 5, Nr. 12 (Dezember 2014): e1567-e1567. http://dx.doi.org/10.1038/cddis.2014.521.

Der volle Inhalt der Quelle
Annotation:
Abstract Adult human adipose-derived mesenchymal stem cells (hAMSCs) are multipotent cells, which are abundant, easily collected, and bypass the ethical concerns that plague embryonic stem cells. Their utility and accessibility have led to the rapid development of clinical investigations to explore their autologous and allogeneic cellular-based regenerative potential, tissue preservation capabilities, anti-inflammatory properties, and anticancer properties, among others. hAMSCs are typically cultured under ambient conditions with 21% oxygen. However, physiologically, hAMSCs exist in an environment of much lower oxygen tension. Furthermore, hAMSCs cultured in standard conditions have shown limited proliferative and migratory capabilities, as well as limited viability. This study investigated the effects hypoxic culture conditions have on primary intraoperatively derived hAMSCs. hAMSCs cultured under hypoxia (hAMSCs-H) remained multipotent, capable of differentiation into osteogenic, chondrogenic, and adipogenic lineages. In addition, hAMSCs-H grew faster and exhibited less cell death. Furthermore, hAMSCs-H had greater motility than normoxia-cultured hAMSCs and exhibited greater homing ability to glioblastoma (GBM) derived from brain tumor-initiating cells from our patients in vitro and in vivo. Importantly, hAMSCs-H did not transform into tumor-associated fibroblasts in vitro and were not tumorigenic in vivo. Rather, hAMSCs-H promoted the differentiation of brain cancer cells in vitro and in vivo. These findings suggest an alternative culturing technique that can enhance the function of hAMSCs, which may be necessary for their use in the treatment of various pathologies including stroke, myocardial infarction, amyotrophic lateral sclerosis, and GBM.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
21

Li, Yuwan, Ziming Liu, Yaping Tang, Qinghong Fan, Wei Feng, Changqi Luo, Guangming Dai et al. „Three-dimensional silk fibroin scaffolds enhance the bone formation and angiogenic differentiation of human amniotic mesenchymal stem cells: a biocompatibility analysis“. Acta Biochimica et Biophysica Sinica 52, Nr. 6 (11.05.2020): 590–602. http://dx.doi.org/10.1093/abbs/gmaa042.

Der volle Inhalt der Quelle
Annotation:
Abstract Silk fibroin (SF) is a fibrous protein with unique mechanical properties, adjustable biodegradation, and the potential to drive differentiation of mesenchymal stem cells (MSCs) along the osteogenic lineage, making SF a promising scaffold material for bone tissue engineering. In this study, hAMSCs were isolated by enzyme digestion and identified by multiple-lineage differentiation. SF scaffold was fabricated by freeze-drying, and the adhesion and proliferation abilities of hAMSCs on scaffolds were determined. Osteoblast differentiation and angiogenesis of hAMSCs on scaffolds were further evaluated, and histological staining of calvarial defects was performed to examine the cocultured scaffolds. We found that hAMSCs expressed the basic surface markers of MSCs. Collagen type I (COL-I) expression was observed on scaffolds cocultured with hAMSCs. The scaffolds potentiated the proliferation of hAMSCs and increased the expression of COL-I in hAMSCs. The scaffolds also enhanced the alkaline phosphatase activity and bone mineralization, and upregulated the expressions of osteogenic-related factors in vitro. The scaffolds also enhanced the angiogenic differentiation of hAMSCs. The cocultured scaffolds increased bone formation in treating critical calvarial defects in mice. This study first demonstrated that the application of 3D SF scaffolds co-cultured with hAMSCs greatly enhanced osteogenic differentiation and angiogenesis of hAMSCs in vitro and in vivo. Thus, 3D SF scaffolds cocultured with hAMSCs may be a better alternative for bone tissue engineering.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
22

Topoluk, Natasha, Richard Hawkins, John Tokish und Jeremy Mercuri. „Amniotic Mesenchymal Stromal Cells Exhibit Preferential Osteogenic and Chondrogenic Differentiation and Enhanced Matrix Production Compared With Adipose Mesenchymal Stromal Cells“. American Journal of Sports Medicine 45, Nr. 11 (25.05.2017): 2637–46. http://dx.doi.org/10.1177/0363546517706138.

Der volle Inhalt der Quelle
Annotation:
Background: Therapeutic efficacy of various mesenchymal stromal cell (MSC) types for orthopaedic applications is currently being investigated. While the concept of MSC therapy is well grounded in the basic science of healing and regeneration, little is known about individual MSC populations in terms of their propensity to promote the repair and/or regeneration of specific musculoskeletal tissues. Two promising MSC sources, adipose and amnion, have each demonstrated differentiation and extracellular matrix (ECM) production in the setting of musculoskeletal tissue regeneration. However, no study to date has directly compared the differentiation potential of these 2 MSC populations. Purpose: To compare the ability of human adipose- and amnion-derived MSCs to undergo osteogenic and chondrogenic differentiation. Study Design: Controlled laboratory study. Methods: MSC populations from the human term amnion were quantified and characterized via cell counting, histologic assessment, and flow cytometry. Differentiation of these cells in comparison to commercially purchased human adipose-derived mesenchymal stromal cells (hADSCs) in the presence and absence of differentiation media was evaluated via reverse transcription polymerase chain reaction (PCR) for bone and cartilage gene transcript markers and histology/immunohistochemistry to examine ECM production. Analysis of variance and paired t tests were performed to compare results across all cell groups investigated. Results: The authors confirmed that the human term amnion contains 2 primary cell types demonstrating MSC characteristics—(1) human amniotic epithelial cells (hAECs) and (2) human amniotic mesenchymal stromal cells (hAMSCs)—and each exhibited more than 90% staining for MSC surface markers (CD90, CD105, CD73). Average viable hAEC and hAMSC yields at harvest were 2.3 × 106 ± 3.7 × 105 and 1.6 × 106 ± 4.7 × 105 per milliliter of amnion, respectively. As well, hAECs and hAMSCs demonstrated significantly greater osteocalcin ( P = .025), aggrecan ( P < .0001), and collagen type 2 ( P = .044) gene expression compared with hADSCs, respectively, after culture in differentiation medium. Moreover, both hAECs and hAMSCs produced significantly greater quantities of mineralized ( P < .0001) and cartilaginous ( P = .0004) matrix at earlier time points compared with hADSCs when cultured under identical osteogenic and chondrogenic differentiation conditions, respectively. Conclusion: Amnion-derived MSCs demonstrate a greater differentiation potential toward bone and cartilage compared with hADSCs. Clinical Relevance: Amniotic MSCs may be the source of choice in the regenerative treatment of bone or osteochondral musculoskeletal disease. They show significantly higher yields and better differentiation toward these tissues than MSCs derived from adipose.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
23

Wu, Shuhong, Zhili Xiao, Jinlin Song, Min Li und Wenhua Li. „Evaluation of BMP-2 Enhances the Osteoblast Differentiation of Human Amnion Mesenchymal Stem Cells Seeded on Nano-Hydroxyapatite/Collagen/Poly(l-Lactide)“. International Journal of Molecular Sciences 19, Nr. 8 (25.07.2018): 2171. http://dx.doi.org/10.3390/ijms19082171.

Der volle Inhalt der Quelle
Annotation:
Background: The aim of this study is to evaluate the effects of recombinant human bone morphogenetic protein 2 (rhBMP-2), human amnion mesenchymal stem cells (hAMSCs), and nanohydroxyapatite/collagen/poly(l-lactide) (nHAC/PLA) in tissue engineering to provide potential approaches for periodontal bone regeneration. Methods: hAMSCs were isolated from discarded amniotic membrane samples and cultured in vitro. Alkaline phosphatase (ALP) staining and alizarin red staining were performed to evaluate the osteoblast (OB) differentiation ability of hAMSCs. Three groups were divided: the experimental group (cells transfected with pcDNA3.1-rhBMP-2), the blank group (cells without gene transfection), and the control group (cells transfected with empty plasmid). RT-PCR and western blot were used to examine whether rhBMP-2 has been successfully expressed. 3-(4,5)-dimethylthiahiazol(-z-y1)-3,5-di-phenytetrazo-liumromide assay (MTT) was done to detect the effect of rhBMP-2 on hAMSCs seeded on nHAC/PLA. ALP activity, mineral formation assay, calcium, phosphate and osteocalcin (OCN) content, and OCN and RUNX2 expression of hAMSCs were detected to evaluate osteogenic differentiation capability of rhBMP-2 on hAMSCs seeded on nHAC/PLA. Results: hAMSCs exhibited intense ALP staining, obvious calcium deposition, and mineralization nodules, and rhBMP-2 were highly expressed in the experimental group. The proliferation of the hAMSCs with rhBMP-2 on nHAC/PLA was significantly higher than the cells without rhBMP-2, and the cells all increased in a time-dependent manner. rhBMP-2 significantly increased the OCN and phosphate content, mineral formation, ALP activity, osteogenic biomarkers OCN, and Runx2, and decreased calcium content in hAMSCs seeded on the nHAC/PLA scaffold. Conclusions: This finding demonstrated that hAMSCs has an ideal OB differentiation ability. rhBMP-2 facilitates the proliferation and osteogenesis of hAMSCs. The nHAC/PLA could act as a good scaffold for hAMSCs seeding, proliferation, and osteogenic differentiation. The application of rhBMP-2, nHAC/PLA, and hAMSCs in tissue engineering may offer promising possibilities for periodontal bone regeneration.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
24

Efrina, Eti, und Nadia Parastama. „ANALISIS PESAN DAKWAH PADA TABLIGH MUSIBAH DALAM CHANNEL USTADZ JUNAIDI HAMSYAH“. JOISCOM (Journal of Islamic Communication) 3, Nr. 1 (30.04.2022): 19–24. http://dx.doi.org/10.36085/joiscom.v3i1.3223.

Der volle Inhalt der Quelle
Annotation:
Dakwah merupakan isi pesan atau materi yang disampaikan da’i kepada mad’u. Pesan dakwah menjadi unsur yang sangat penting dalam menyampaikan dakwah dan sangat menentukan keberhasilan dakwah itu sendiri. Suatu kegiatan dakwah yang dilaksanakan dikatakan efektif apabila materinya bersifat informatif, edukatif, dan solusif bagi masyarakat.Adapun rumusan masalah pada penelitian ini bertujuan untuk mengetahui bagaimana analisis pesan dakwah pada tabligh musibah dalam channel youtube Ustadz Junaidi Hamsyah, dan untuk mengetahui metode pengemasan dan penyampaian pesan-pesan dakwah.Penelitian ini dilakukan pada channel youtube Ustadz Junaidi Hamsyah dengan cara menganalisis isi pesan dakwah yang terdapat di 10 vidio dengan tema tabligh musibah. Jenis penelitian ini adalah analisis isi (content analisis) terhadap isi pesan dakwah pada channel youtube Ustadz Junaidi Hamsyah. Bahan yang dipelajari dapat berupa bahan yang diucapkan dan bahan yang ditulis. Pengertian lain menyebutkan bahwa analisis isi (content analysis) adalah teknik penelitian untuk membuat referensi-referensi yang dapat ditiru (replicable), dan shahih dengan memerhatikan konteksnya. Analisis ini berhubungan dengan komunikasi atau isi komunikasi.Berdasarkan hasil analisis yang dilakukan pada channel youtube Ustadz Junaidi Hamsyah dapat disimpulkan bahwa metode pengemasan  dan penyampaian dakwah yang digunakan oleh ustadz junaidi hamsyah adalah jenis metode Al Mau’idzatil Hasanah (pelajaran yang baik), dan pesan yang disampaikan dalam 10 vidio yang di analisis adalah mencakup pesan aqidah, pesan syariah dan pesan akhlak.Kata kunci : Channel Youtube, Content Analisys, Dakwah, Ustadz junaidi Hamsyah
APA, Harvard, Vancouver, ISO und andere Zitierweisen
25

Bispo, Daniela S. C., Lenka Michálková, Marlene Correia, Catarina S. H. Jesus, Iola F. Duarte, Brian J. Goodfellow, Mariana B. Oliveira, João F. Mano und Ana M. Gil. „Endo- and Exometabolome Crosstalk in Mesenchymal Stem Cells Undergoing Osteogenic Differentiation“. Cells 11, Nr. 8 (07.04.2022): 1257. http://dx.doi.org/10.3390/cells11081257.

Der volle Inhalt der Quelle
Annotation:
This paper describes, for the first time to our knowledge, a lipidome and exometabolome characterization of osteogenic differentiation for human adipose tissue stem cells (hAMSCs) using nuclear magnetic resonance (NMR) spectroscopy. The holistic nature of NMR enabled the time-course evolution of cholesterol, mono- and polyunsaturated fatty acids (including ω-6 and ω-3 fatty acids), several phospholipids (phosphatidylcholine, phosphatidylethanolamine, sphingomyelins, and plasmalogens), and mono- and triglycerides to be followed. Lipid changes occurred almost exclusively between days 1 and 7, followed by a tendency for lipidome stabilization after day 7. On average, phospholipids and longer and more unsaturated fatty acids increased up to day 7, probably related to plasma membrane fluidity. Articulation of lipidome changes with previously reported polar endometabolome profiling and with exometabolome changes reported here in the same cells, enabled important correlations to be established during hAMSC osteogenic differentiation. Our results supported hypotheses related to the dynamics of membrane remodelling, anti-oxidative mechanisms, protein synthesis, and energy metabolism. Importantly, the observation of specific up-taken or excreted metabolites paves the way for the identification of potential osteoinductive metabolites useful for optimized osteogenic protocols.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
26

Hendrijantini, Nike, Poedjo Hartono, Helen Susilowati, Cindy K. Hartono, Reni P. Daniati und Febrian Brahmana. „Study of Human Amniotic Membrane Mesenchymal Stem Cells Using Gelatin and Alginate as Nontoxic Scaffolds“. Recent Advances in Biology and Medicine 5 (2019): 1. http://dx.doi.org/10.18639/rabm.2019.877306.

Der volle Inhalt der Quelle
Annotation:
Perinatal mesenchymal stem cells (MSCs), for example, from amniotic membrane, have advantages over adult sources, such as bone marrow, in terms of ease of availability, cell naivety, tissue stem cell abundance, high capacity of proliferation, and less donor site morbidity, because it does not require invasive procedures. Natural polymer scaffolds, such as gelatin and alginate, are biocompatible. Combination of stem cells from amniotic membrane (hAMSCs) and gelatin or alginate as scaffold can be promising. However, cytotoxicity comparison of gelatin and alginate to hAMSCs has not been widely studied. This study was aimed to compare cytotoxicity of gelatin and alginate on hAMSCs in vitro. Isolation and culture were performed on hAMSCs of the healthy full-term pregnancy. In passage 4, Flow Cytometry CD90, CD105, and CD73 phenotype characterization was done. Colorimetric assay of 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) was performed to measure the cytotoxicity. There were three sample groups: (control group) hAMSCs with alpha-minimum essential medium (α-MEM) solution as control; (gelatin group) hAMSCs with gelatin; (alginate group) hAMSCs with alginate. Each group consisted of 12 samples. Flow cytometry of hAMSCs expressed 28.78% CD90, 36.95% CD105, and 44.41% CD73 surface markers. No sample depicted toxicity in either gelatin or alginate group, and this is indicated by the average percentage of living cells in gelatin 97.26% and in alginate 98.43%. No statistically significant difference (ρ=0.057) of cytotoxicity was found between gelatin and alginate to hAMSCs. Gelatin and alginate were nontoxic to hAMSCs in vitro.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
27

Wang, Yuli, Fei Jiang, Yi Liang, Ming Shen und Ning Chen. „Human Amnion-Derived Mesenchymal Stem Cells Promote Osteogenic Differentiation in Human Bone Marrow Mesenchymal Stem Cells by Influencing the ERK1/2 Signaling Pathway“. Stem Cells International 2016 (2016): 1–12. http://dx.doi.org/10.1155/2016/4851081.

Der volle Inhalt der Quelle
Annotation:
Human amnion-derived mesenchymal stem cells (HAMSCs) are considered to be an important resource in the field of tissue engineering because of their anti-inflammatory properties and fewer ethical issues associated with their use compared with other sources of stem cells. HAMSCs can be obtained from human amniotic membranes, a readily available and abundant tissue. However, the potential of HAMSCs as seed cells for treating bone deficiency is unknown. In this study, HAMSCs were used to promote proliferation and osteoblastic differentiation in human bone marrow mesenchymal stem cells (HBMSCs) in a Transwell coculture system. Proliferation levels were investigated by flow cytometry and immunofluorescence staining of 5-ethynyl-2′-deoxyuridine (EdU). Osteoblastic differentiation and mineralization were evaluated in chromogenic alkaline phosphatase (ALP) activity substrate assays, Alizarin red S staining, and RT-PCR analysis of early HBMSCs osteogenic marker expression. We demonstrated that HAMSCs stimulated increased alkaline phosphatase (ALP) activity, mRNA expression of osteogenic marker genes, and mineralized matrix deposition. Moreover, the effect of HAMSCs was significantly inhibited by U0126, a highly selective inhibitor of extracellular signaling-regulated kinase 1/2 (ERK1/2) signaling. We demonstrate that HAMSCs promote osteogenic differentiation in HBMSCs by influencing the ERK1/2 signaling pathway. These observations confirm the potential of HAMSCs as a seed cell for the treatment of bone deficiency.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
28

Xiao, Shune, Guangtao Huang, Zairong Wei, Kaiyu Nie, Zhiyuan Liu, Chengliang Deng und Dali Wang. „IL-10 Gene-Modified Human Amniotic Mesenchymal Stem Cells Augment Regenerative Wound Healing by Multiple Synergistic Effects“. Stem Cells International 2019 (11.06.2019): 1–13. http://dx.doi.org/10.1155/2019/9158016.

Der volle Inhalt der Quelle
Annotation:
Mesenchymal stem cells (MSCs) possess a capacity to enhance cutaneous wound healing that is well characterized. However, the therapeutic effect of MSCs appears to be limited. Modifying MSC target genes to increase necessary biological effects is a promising strategy for wound therapy. Interleukin-10 (IL-10) is an anti-inflammatory cytokine that has a therapeutic effect on wound healing. In this study, we modified human amniotic mesenchymal stem cells (hAMSCs) using recombinant lentiviral vectors for expressing IL-10 and evaluated the therapeutic effects of hAMSCs-IL-10 in wound healing. We elucidated the mechanisms underlying the effects. We found that promoting wound healing was maintained by synergistic effects of hAMSCs and IL-10. hAMSCs-IL-10 showed stronger biological effects in accelerating wound closure, enhancing angiogenesis, modulating inflammation, and regulating extracellular matrix remodeling than hAMSCs. hAMSCs-IL-10 would be better at promoting wound healing and improving healing quality. These data may provide a theoretical foundation for clinical administration of hAMSCs-IL-10 in cutaneous wound healing and skin regeneration.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
29

Chen, Tzu-Jou, Yen-Ting Yeh, Fu-Shiang Peng, Ai-Hsien Li und Shinn-Chih Wu. „S100A8/A9 Enhances Immunomodulatory and Tissue-Repairing Properties of Human Amniotic Mesenchymal Stem Cells in Myocardial Ischemia-Reperfusion Injury“. International Journal of Molecular Sciences 22, Nr. 20 (16.10.2021): 11175. http://dx.doi.org/10.3390/ijms222011175.

Der volle Inhalt der Quelle
Annotation:
Paracrine factors of human mesenchymal stem cells (hMSCs) have the potential of preventing adverse cardiac remodeling after myocardial infarction (MI). S100A8 and S100A9 are calcium-binding proteins playing essential roles in the regulation of inflammation and fibrous tissue formation, and they might modulate the paracrine effect of hMSCs. We isolated human amniotic mesenchymal stem cells (hAMSCs) and examined the changes in the expression level of regulatory genes of inflammation and fibrosis after hAMSCs were treated with S100A8/A9. The anti-inflammatory and anti-fibrotic effects of hAMSCs pretreated with S100A8/A9 were shown to be superior to those of hAMSCs without S100A8/A9 pretreatment in the cardiomyocyte hypoxia/reoxygenation experiment. We established a murine myocardial ischemia/reperfusion model to compare the therapeutic effects of the conditioned medium of hAMSCs with or without S100A8/A9 pretreatment. We found the hearts administered with a conditioned medium of hAMSCs with S100A8/A9 pretreatment had better left ventricular systolic function on day 7, 14, and 28 after MI. These results suggest S100A8/A9 enhances the paracrine therapeutic effects of hAMSCs in aspects of anti-inflammation, anti-fibrosis, and cardiac function preservation after MI.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
30

Zhang, Jun, Ziming Liu, Yuwan Li, Qi You, Jibin Yang, Ying Jin, Gang Zou, Jingfeng Tang, Zhen Ge und Yi Liu. „FGF-2-Induced Human Amniotic Mesenchymal Stem Cells Seeded on a Human Acellular Amniotic Membrane Scaffold Accelerated Tendon-to-Bone Healing in a Rabbit Extra-Articular Model“. Stem Cells International 2020 (06.01.2020): 1–14. http://dx.doi.org/10.1155/2020/4701476.

Der volle Inhalt der Quelle
Annotation:
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.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
31

Matteo, Beccia, Carbone, Castellani, Milillo, Lauritano, Gioia, Angiolillo und Conese. „Effect of Mother’s Age and Pathology on Functional Behavior of Amniotic Mesenchymal Stromal Cells—Hints for Bone Regeneration“. Applied Sciences 9, Nr. 17 (22.08.2019): 3471. http://dx.doi.org/10.3390/app9173471.

Der volle Inhalt der Quelle
Annotation:
Human amnion-derived mesenchymal stromal cells (hAMSCs) are used increasingly in regenerative medicine applications, including dentistry. The aim of this study was to evaluate if hAMSCs from aged and pathological mothers could be affected in their phenotype and functional behavior. hAMSCs were isolated from placentas of women aged younger than 40 years (Group 1, n = 7), older than 40 years (Group 2, n = 6), and with pre-eclampsia (Group 3, n = 5). Cell yield and viability were assessed at isolation (p0). Cell proliferation was evaluated from p0 to p5. Passage 2 was used to determine the phenotype, the differentiation capacity, and the adhesion to machined and sandblasted titanium disks. hAMSCs recovered from Group 3 were fewer than in Group 1. Viability and doubling time were not different among the three groups. Percentages of CD29+ cells were significantly lower in Group 3, while percentages of CD73+ cells were significantly lower in Groups 2 and 3 as compared with Group 1. hAMSCs from Group 2 showed a significant lower differentiation capacity towards chondrogenic and osteogenic lineages. hAMSCs from Group 3 adhered less to titanium surfaces. In conclusion, pathology can affect hAMSCs in phenotype and functional behavior and may alter bone regeneration capacities.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
32

Ledda, Mario, Enrico D’Emilia, Maria Lolli, Rodolfo Marchese, Claudio De Lazzari und Antonella Lisi. „Non-Ionizing Radiation for Cardiac Human Amniotic Mesenchymal Stromal Cell Commitment: A Physical Strategy in Regenerative Medicine“. International Journal of Molecular Sciences 19, Nr. 8 (08.08.2018): 2324. http://dx.doi.org/10.3390/ijms19082324.

Der volle Inhalt der Quelle
Annotation:
Cell therapy is an innovative strategy for tissue repair, since adult stem cells could have limited regenerative ability as in the case of myocardial damage. This leads to a local contractile dysfunction due to scar formation. For these reasons, refining strategy approaches for “in vitro” stem cell commitment, preparatory to the “in vivo” stem cell differentiation, is imperative. In this work, we isolated and characterized at molecular and cellular level, human Amniotic Mesenchymal Stromal Cells (hAMSCs) and exposed them to a physical Extremely Low Frequency Electromagnetic Field (ELF-EMF) stimulus and to a chemical Nitric Oxide treatment. Physically exposed cells showed a decrease of cell proliferation and no change in metabolic activity, cell vitality and apoptotic rate. An increase in the mRNA expression of cardiac and angiogenic differentiation markers, confirmed at the translational level, was also highlighted in exposed cells. Our data, for the first time, provide evidence that physical ELF-EMF stimulus (7 Hz, 2.5 µT), similarly to the chemical treatment, is able to trigger hAMSC cardiac commitment. More importantly, we also observed that only the physical stimulus is able to induce both types of commitments contemporarily (cardiac and angiogenic), suggesting its potential use to obtain a better regenerative response in cell-therapy protocols.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
33

Paracchini, Valentina, Annalucia Carbone, Federico Colombo, Stefano Castellani, Silvia Mazzucchelli, Sante Di Gioia, Dario Degiorgio et al. „Amniotic Mesenchymal Stem Cells: A New Source for Hepatocyte-Like Cells and Induction of CFTR Expression by Coculture with Cystic Fibrosis Airway Epithelial Cells“. Journal of Biomedicine and Biotechnology 2012 (2012): 1–15. http://dx.doi.org/10.1155/2012/575471.

Der volle Inhalt der Quelle
Annotation:
Cystic fibrosis (CF) is a monogenic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, with lung and liver manifestations. Because of pitfalls of gene therapy, novel approaches for reconstitution of the airway epithelium and CFTR expression should be explored. In the present study, human amniotic mesenchymal stem cells (hAMSCs) were isolated from term placentas and characterized for expression of phenotypic and pluripotency markers, and for differentiation potential towards mesoderm (osteogenic and adipogenic) lineages. Moreover, hAMSCs were induced to differentiate into hepatocyte-like cells, as demonstrated by mixed function oxidase activity and expression of albumin, alpha1-antitrypsin, and CK19. We also investigated the CFTR expression in hAMSCs upon isolation and in coculture with CF airway epithelial cells. Freshly isolated hAMSCs displayed low levels of CFTR mRNA, which even decreased with culture passages. Following staining with the vital dye CM-DiI, hAMSCs were mixed with CFBE41o- respiratory epithelial cells and seeded onto permeable filters. Flow cytometry demonstrated that 33–50% of hAMSCs acquired a detectable CFTR expression on the apical membrane, a result confirmed by confocal microscopy. Our data show that amniotic MSCs have the potential to differentiate into epithelial cells of organs relevant in CF pathogenesis and may contribute to partial correction of the CF phenotype.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
34

Li, Yuwan, Ziming Liu, Ying Jin, Xizhong Zhu, Shengmin Wang, Jibin Yang, Youliang Ren et al. „Differentiation of Human Amniotic Mesenchymal Stem Cells into Human Anterior Cruciate Ligament Fibroblast Cells by In Vitro Coculture“. BioMed Research International 2017 (2017): 1–15. http://dx.doi.org/10.1155/2017/7360354.

Der volle Inhalt der Quelle
Annotation:
Anterior cruciate ligament injuries are common in humans, though cellular components of the knee have little regenerative or proliferation potential. This study investigated the differentiation of human amnion-derived mesenchymal stem cells (hAMSCs) into human anterior cruciate ligament fibroblasts (hACLFs) in vitro through induction with bFGF and TGF-β1 with coculture systems. Groups A and B comprised hAMSCs at the 3rd passage cultured with and without bFGF and TGF-β1, respectively; Groups C and D consisted of hAMSCs and hACLFs in monolayer coculture with and without bFGF and TGF-β1, respectively; Groups E and F were composed of hAMSCs and hACLFs in Transwell coculture with and without bFGF and TGF-β1, respectively. Cell morphology and proliferation were recorded. Protein expression and relative mRNA expression were evaluated in each group. Cell proliferation was significantly higher in the induced groups than in the noninduced groups. Protein expression increased over time with the highest expression observed in Group E. mRNA levels were significantly higher in Group E than in other groups. This study is the first to demonstrate the use of the Transwell coculture system for this purpose, and hAMSCs were successfully differentiated into hACLFs. Thus, hAMSCs may be a superior choice for hACLF differentiation via Transwell coculture.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
35

Riedel, Rodrigo, Soledad Pérez-Amodio, Laura Cabo-Zabala, Eugenio Velasco-Ortega, Julieta Maymó, Javier Gil, Loreto Monsalve-Guil et al. „Influence of the Surface Topography of Titanium Dental Implants on the Behavior of Human Amniotic Stem Cells“. International Journal of Molecular Sciences 25, Nr. 13 (06.07.2024): 7416. http://dx.doi.org/10.3390/ijms25137416.

Der volle Inhalt der Quelle
Annotation:
The dental implant surface plays a crucial role in osseointegration. The topography and physicochemical properties will affect the cellular functions. In this research, four distinct titanium surfaces have been studied: machined acting (MACH), acid etched (AE), grit blasting (GBLAST), and a combination of grit blasting and subsequent acid etching (GBLAST + AE). Human amniotic mesenchymal (hAMSCs) and epithelial stem cells (hAECs) isolated from the amniotic membrane have attractive stem-cell properties. They were cultured on titanium surfaces to analyze their impact on biological behavior. The surface roughness, microhardness, wettability, and surface energy were analyzed using interferometric microscopy, Vickers indentation, and drop-sessile techniques. The GBLAST and GBLAST + AE surfaces showed higher roughness, reduced hydrophilicity, and lower surface energy with significant differences. Increased microhardness values for GBLAST and GBLAST + AE implants were attributed to surface compression. Cell viability was higher for hAMSCs, particularly on GBLAST and GBLAST + AE surfaces. Alkaline phosphatase activity enhanced in hAMSCs cultured on GBLAST and GBLAST + AE surfaces, while hAECs showed no mineralization signals. Osteogenic gene expression was upregulated in hAMSCs on GBLAST surfaces. Moreover, α2 and β1 integrin expression enhanced in hAMSCs, suggesting a surface−integrin interaction. Consequently, hAMSCs would tend toward osteoblastic differentiation on grit-blasted surfaces conducive to osseointegration, a phenomenon not observed in hAECs.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
36

Gao, Ya, Ying Xu, Weiru Li, Yintian Zhang, Baohong Ping und Qifa Liu. „Effects of Interferon-γ on Proliferation and Ability of Secretion of Human Amniotic Mesenchymal Stem Cells“. Blood 134, Supplement_1 (13.11.2019): 5012. http://dx.doi.org/10.1182/blood-2019-125500.

Der volle Inhalt der Quelle
Annotation:
Objective:The immunoregulatory properties and proliferation of mesenchymal stem cells (MSCs) could be affected by inflammatory factors. However, there have been few studies about human amniotic MSCs (hAMSCs). We investigated the effects of interferon (IFN)-γ on the proliferation and apoptosis of hAMSCs, and measured the level of inflammatory factors secreted by hAMSCs. Result:hAMSCs were cultured with complete medium with different concentrations of IFN-γ. We detected the proliferation of hAMSCs by Cell Counting Kit-8 assays, analysed apoptosis by flow cytometry (FCM) at 48 h, and mesasured the level of inflammatory factors such as solube HLA-G and prostaglandin E2 (PGE2) in the supernatant at 48 h by ELISA. The level of kynurenine (KYN) was measured by ultraviolet spectrophotometry. As culture time increased, the proliferation of hAMSCs with different concentrations of IFN-γ increased rapidly from day 1 to day 4, and then the growth rate slowed. FCM indicated that there was no significant apoptosis in the 100 ng/ml IFN-γ group compared with cells without IFN-γ. The level of PGE2 and soluble HLA-G in cells with IFN-γ was higher compared with those without IFN-γ. The level of KYN increased significantly in the cells with IFN-γ. Conclusion:IFN-γ did not affect the growth and proliferation of hAMSCs, and promoted secretion of PGE2 and soluble HLA-G, and enhanced activity of indoleamine 2,3-dioxygenase (IDO), providing a theoretical basis for hAMSCs to prevent and treat graft-versus-host disease. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
37

Gorjipour, Fazel, Ladan Hosseini-Gohari, Alireza Alizadeh Ghavidel, Seyed Javad Hajimiresmaiel, Nasim Naderi, Amir Darbandi Azar und Hamidreza Pazoki-Toroudi. „Mesenchymal stem cells from human amniotic membrane differentiate into cardiomyocytes and endothelial-like cells without improving cardiac function after surgical administration in rat model of chronic heart failure“. Journal of Cardiovascular and Thoracic Research 11, Nr. 1 (25.02.2019): 35–42. http://dx.doi.org/10.15171/jcvtr.2019.06.

Der volle Inhalt der Quelle
Annotation:
Introduction: Human amnion-derived mesenchymal stem cells (hAMSCs) have been used in the treatment of acute myocardial infarction. In the current study, we investigated the efficacy of hAMSCs for the treatment of chronic model of myocardial ischemia and heart failure (HF) in rats. Methods: Male Wistar rats weighing between 250 to 350 g were randomized into three groups: sham, HF control and HF+hAMSCs. For HF induction, animals were anesthetized and underwent left anterior descending artery ligation. In HF+hAMSCs group, 2×106 cells were injected into the left ventricular muscle four weeks post ischemia in the border zone of the ischemic area. Cardiac function was studied using echocardiography. Masson’s trichrome staining was used for studying tissue fibrosis. Cells were transduced with green fluorescent protein (GFP) coding lentiviral vector. Immunohistochemistry was used for detecting GFP, vascular-endothelial growth factor (VEGF) and troponin T markers in the tissue sections. Results: Assessment of the cardiac function revealed no improvement in the myocardial function compared to the control HF group. Moreover, tissue fibrosis was similar in two groups. Immunohistochemical study revealed the homing of the injected hAMSCs to the myocardium. Cells were stained positive for VEGF and troponin T markers. Conclusion: injection of hAMSCs 4 weeks after ischemia does not improve cardiac function and cardiac muscle fibrosis, although the cells show markers of differentiation into vascular endothelial cells and cardiomyocytes. In sum, it appears that hAMSCs are effective in the early phases of myocardial ischemia and does not offer a significant advantage in patients with chronic HF.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
38

Beccia, Elisa, Valeria Daniello, Onofrio Laselva, Giorgia Leccese, Michele Mangiacotti, Sante Di Gioia, Gianfranco La Bella et al. „Human Amniotic Mesenchymal Stem Cells and Fibroblasts Accelerate Wound Repair of Cystic Fibrosis Epithelium“. Life 12, Nr. 5 (19.05.2022): 756. http://dx.doi.org/10.3390/life12050756.

Der volle Inhalt der Quelle
Annotation:
Cystic fibrosis (CF) airways are affected by a deranged repair of the damaged epithelium resulting in altered regeneration and differentiation. Previously, we showed that human amniotic mesenchymal stem cells (hAMSCs) corrected base defects of CF airway epithelial cells via connexin (CX)43-intercellular gap junction formation. In this scenario, it is unknown whether hAMSCs, or fibroblasts sharing some common characteristics with MSCs, can operate a faster repair of a damaged airway epithelium. A tip-based scratch assay was employed to study wound repair in monolayers of CFBE14o- cells (CFBE, homozygous for the F508del mutation). hAMSCs were either co-cultured with CFBE cells before the wound or added to the wounded monolayers. NIH-3T3 fibroblasts (CX43+) were added to wounded cells. HeLa cells (CX43-) were used as controls. γ-irradiation was optimized to block CFBE cell proliferation. A specific siRNA was employed to downregulate CX43 expression in CFBE cells. CFBE cells showed a delayed repair as compared with wt-CFTR cells (16HBE41o-). hAMSCs enhanced the wound repair rate of wounded CFBE cell monolayers, especially when added post wounding. hAMSCs and NIH-3T3 fibroblasts, but not HeLa cells, increased wound closure of irradiated CFBE monolayers. CX43 downregulation accelerated CFBE wound repair rate without affecting cell proliferation. We conclude that hAMSCs and fibroblasts enhance the repair of a wounded CF airway epithelium, likely through a CX43-mediated mechanism mainly involving cell migration.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
39

Silini, Antonietta R., Valentina Spoldi, Silvia De Munari, Elsa Vertua, Fabiola Munarin, Paola Petrini, Silvia Farè und Ornella Parolini. „Immunological and Differentiation Properties of Amniotic Cells Are Retained After Immobilization in Pectin Gel“. Cell Transplantation 27, Nr. 1 (Januar 2018): 70–76. http://dx.doi.org/10.1177/0963689717738786.

Der volle Inhalt der Quelle
Annotation:
Mesenchymal stromal cells from the human amniotic membrane (i.e., human amniotic mesenchymal stromal cells [hAMSCs]) of term placenta are increasingly attracting attention for their applications in regenerative medicine. Osteochondral defects represent a major clinical problem with lifelong chronic pain and compromised quality of life. Great promise for osteochondral regeneration is held in hydrogel-based constructs that have a flexible composition and mimic the physiological structure of cartilage. Cell loading within a hydrogel represents an advantage for regenerative purposes, but the encapsulation steps can modify cell properties. As pectin gels have also been explored as cell vehicles on 3D scaffolds, the aim of this study was to explore the possibility to include hAMSCs in pectin gel. Immobilization of hAMSCs into pectin gels could expand their application in cell-based bioengineering strategies. hAMSCs were analyzed for their viability and recovery from the pectin gel and for their ability to differentiate toward the osteogenic lineage and to maintain their immunological characteristics. When treated with a purposely designed pectin/hydroxyapatite gel biocomposite, hAMSCs retained their ability to differentiate toward the osteogenic lineage, did not induce an immune response, and retained their ability to reduce T cell proliferation. Taken together, these results suggest that hAMSCs could be used in combination to pectin gels for the study of novel osteochondral regeneration strategies.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
40

Luo, Yi, Ai-Tong Wang, Qing-Fang Zhang, Ru-Ming Liu und Jian-Hui Xiao. „RASL11B gene enhances hyaluronic acid-mediated chondrogenic differentiation in human amniotic mesenchymal stem cells via the activation of Sox9/ERK/smad signals“. Experimental Biology and Medicine 245, Nr. 18 (02.09.2020): 1708–21. http://dx.doi.org/10.1177/1535370220944375.

Der volle Inhalt der Quelle
Annotation:
This study aimed to elucidate the molecular mechanisms, whereby hyaluronic acid, a main extracellular matrix component of articular cartilage, promotes the chondrogenic differentiation of human amniotic mesenchymal stem cells (hAMSCs). Our previous findings indicated that hyaluronic acid combined with hAMSCs showed a marked therapeutic effect against rat osteoarthritis. In the present study, hyaluronic acid markedly enhanced the expression of chondrocyte-specific markers including Col2α1, Acan, and Sox9 in hAMSCs, with strong synergistic effects on chondrogenic differentiation, in combination with the commonly used inducer, transforming growth factor β3 (TGF-β3). Microarray analysis showed that Ras-like protein family member 11B (RASL11B) played a pivotal role in the process of hyaluronic acid-mediated chondrogenesis of hAMSCs. This directional differentiation was significantly inhibited by RASL11B knockdown, but RASL11B overexpression dramatically promoted the expression of Sox9, a master chondrogenesis transcriptional factor, at the levels of transcription and translation. Increased Sox9 expression subsequently resulted in high expression levels of Col2α1 and Acan and the accumulation of cartilage-specific matrix components, such as type 2 collagen and glycosaminoglycans. Moreover, we observed that RASL11B activated the signal molecules such as ERK1/2, and Smad2/3 in the presence of hyaluronic acid during TGF-β3-induced chondrogenesis of hAMSCs. Taken together, these findings suggest that hyaluronic acid activates the RASL11B gene to potentiate the chondrogenic differentiation of hAMSCs via the activation of Sox9 and ERK/Smad signaling, thus providing a new strategy for cartilage defect repairing by hyaluronic acid-based stem cell therapy.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
41

Schiffman, Carl. „Curly Hamson Learns How to Eat“. Antioch Review 61, Nr. 2 (2003): 345. http://dx.doi.org/10.2307/4614485.

Der volle Inhalt der Quelle
APA, Harvard, Vancouver, ISO und andere Zitierweisen
42

Schiffman, Carl. „Curly Hamson and the Lucky Couple“. Antioch Review 64, Nr. 2 (2006): 350. http://dx.doi.org/10.2307/4614990.

Der volle Inhalt der Quelle
APA, Harvard, Vancouver, ISO und andere Zitierweisen
43

Qu, Rongmei, Kai He, Tingyu Fan, Yuchao Yang, Liyao Mai, Zhiwei Lian, Zhitao Zhou et al. „Single-Cell Transcriptomic Sequencing Analyses of Cell Heterogeneity During Osteogenesis of Human Adipose-Derived Mesenchymal Stem Cells“. Stem Cells 39, Nr. 11 (16.08.2021): 1478–88. http://dx.doi.org/10.1002/stem.3442.

Der volle Inhalt der Quelle
Annotation:
Abstract Mesenchymal stem cells (MSCs) are known for their multilineage differentiation potential with immune-modulatory properties. The molecular underpinnings of differentiation remain largely undefined. In this study, we investigated the cellular and molecular features of chemically induced osteogenesis from MSC isolated from human adipose tissue (human adipose MSCs, hAMSCs) using single-cell RNA-sequencing (scRNA-seq). We found that a near complete differentiation of osteogenic clusters from hAMSCs under a directional induction. Both groups of cells are heterogeneous, and some of the hAMSCs cells are intrinsically prepared for osteogenesis, while variant OS clusters seems in cooperation with a due division of the general function. We identified a set of genes related to cell stress response highly expressed during the differentiation. We also characterized a series of transitional transcriptional waves throughout the process from hAMSCs to osteoblast and specified the unique gene networks and epigenetic status as key markers of osteogenesis.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
44

Zito, Giovanni, Vitale Miceli, Claudia Carcione, Rosalia Busà, Matteo Bulati, Alessia Gallo, Gioacchin Iannolo, Duilio Pagano und Pier Giulio Conaldi. „Human Amnion-Derived Mesenchymal Stromal/Stem Cells Pre-Conditioning Inhibits Inflammation and Apoptosis of Immune and Parenchymal Cells in an In Vitro Model of Liver Ischemia/Reperfusion“. Cells 11, Nr. 4 (17.02.2022): 709. http://dx.doi.org/10.3390/cells11040709.

Der volle Inhalt der Quelle
Annotation:
Ischemia/reperfusion injury (IRI) represents one of the leading causes of primary non-function acute liver transplantation failure. IRI, generated by an interruption of organ blood flow and the subsequent restoration upon transplant, i.e., reperfusion, generates the activation of an inflammatory cascade from the resident Kupffer cells, leading first to neutrophils recruitment and second to apoptosis of the parenchyma. Recently, human mesenchymal stromal/stem cells (hMSCs) and derivatives have been implemented for reducing the damage induced by IRI. Interestingly, sparse data in the literature have described the use of human amnion-derived MSCs (hAMSCs) and, more importantly, no evidence regarding hMSCs priming on liver IRI have been described yet. Thus, our study focused on the definition of an in vitro model of liver IRI to test the effect of primed hAMSCs to reduce IRI damage on immune and hepatic cells. We found that the IFNγ pre-treatment and 3D culture of hAMSCs strongly reduced inflammation induced by M1-differentiated macrophages. Furthermore, primed hAMSCs significantly inhibited parenchymal apoptosis at early timepoints of reperfusion by blocking the activation of caspase 3/7. All together, these data demonstrate that hAMSCs priming significantly overcomes IRI effects in vitro by engaging the possibility of defining the molecular pathways involved in this process.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
45

Miceli, Vitale, Mariangela Pampalone, Serena Vella, Anna Paola Carreca, Giandomenico Amico und Pier Giulio Conaldi. „Comparison of Immunosuppressive and Angiogenic Properties of Human Amnion-Derived Mesenchymal Stem Cells between 2D and 3D Culture Systems“. Stem Cells International 2019 (18.02.2019): 1–16. http://dx.doi.org/10.1155/2019/7486279.

Der volle Inhalt der Quelle
Annotation:
The secretion of potential therapeutic factors by mesenchymal stem cells (MSCs) has aroused much interest given the benefits that it can bring in the field of regenerative medicine. Indeed, the in vitro multipotency of these cells and the secretive capacity of both angiogenic and immunomodulatory factors suggest a role in tissue repair and regeneration. However, during culture, MSCs rapidly lose the expression of key transcription factors associated with multipotency and self-renewal, as well as the ability to produce functional paracrine factors. In our study, we show that a three-dimensional (3D) culture method is effective to induce MSC spheroid formation, to maintain the multipotency and to improve the paracrine activity of a specific population of human amnion-derived MSCs (hAMSCs). The regenerative potential of both 3D culture-derived conditioned medium (3D CM) and their exosomes (EXO) was assessed against 2D culture products. In particular, tubulogenesis assays revealed increased capillary maturation in the presence of 3D CM compared with both 2D CM and 2D EXO. Furthermore, 3D CM had a greater effect on inhibition of PBMC proliferation than both 2D CM and 2D EXO. To support this data, hAMSC spheroids kept in our 3D culture system remained viable and multipotent and secreted considerable amounts of both angiogenic and immunosuppressive factors, which were detected at lower levels in 2D cultures. This work reveals the placenta as an important source of MSCs that can be used for eventual clinical applications as cell-free therapies.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
46

Bounoure, Gilles. „Art of the Boiken de Michael Hamson“. Journal de la société des océanistes, Nr. 135 (31.12.2012): 273–75. http://dx.doi.org/10.4000/jso.6742.

Der volle Inhalt der Quelle
APA, Harvard, Vancouver, ISO und andere Zitierweisen
47

Li, Man, Ji Li, Hong Chen und Mingxin Zhu. „VEGF-Expressing Mesenchymal Stem Cell Therapy for Safe and Effective Treatment of Pain in Parkinson’s Disease“. Cell Transplantation 32 (Januar 2023): 096368972211491. http://dx.doi.org/10.1177/09636897221149130.

Der volle Inhalt der Quelle
Annotation:
Vascular endothelial growth factor (VEGF) is a pro-angiogenic factor that mediates the differentiation and function of vascular endothelial cells. VEGF has been implicated in modulating various pains. However, the effects of VEGF in Parkinson’s disease (PD)-related pain have not been studied. The goal of this study was to understand the effects of VEGF-expressing mesenchymal stem cells (MSCs) on PD-related pain and the involved mechanisms. We used two types of MSCs: hAMSC-Vector-GFP and hAMSC-VEGF189-GFP in PD mice. Then, the expression of VEGF and the viability have been compared between two types of MSCs. To demonstrate the therapeutic effect of hAMSC-VEGF189-GFP, we transplanted each cell line in a PD mouse model. Head mechanical withdrawal thresholds were examined. hAMSC-VEGF189-GFP was associated with significantly increased VEGF expression and slightly increased viability, compared with hAMSC-Vector-GFP. The transplanted hAMSC-VEGF189-GFP significantly improved mechanical allodynia and inhibited transient receptor potential vanilloid 1 (TRPV1) expression in site. And such pain relief effects could be partially blocked by TRPV1 agonist. However, we did not observe tumor generation or neuron degeneration in hAMSC-VEGF189-GFP-transplanted animals. Taken together, our data suggest that hAMSC-VEGF189-GFP is safely therapeutically appropriate for treating PD-related pain. VEGF inhibits TRPV1 expression, which may contribute to its analgesic properties.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
48

Basok, Y. B., A. M. Grigoryev, L. A. Kirsanova, A. D. Kirillova, A. M. Subbot, A. V. Tsvetkova, E. A. Nemets und V. I. Sevastianov. „Comparative study of chondrogenesis of human adipose-derived mesenchymal stem cells when cultured in collagen-containing media under in vitro conditions“. Russian Journal of Transplantology and Artificial Organs 23, Nr. 3 (16.09.2021): 90–100. http://dx.doi.org/10.15825/1995-1191-2021-3-90-100.

Der volle Inhalt der Quelle
Annotation:
In terms of method of production, collagen carriers are subdivided into materials obtained on the basis of extracellular matrix (ECM) components, particularly collagen-containing hydrogels and decellularized tissue.Objective: to compare in vitro the ability of biopolymer microheterogeneous collagen-containing hydrogel (BMCH) and tissue-specific matrix from decellularized porcine articular cartilage (DPAC) to support adhesion, proliferation and chondrogenic differentiation of human adipose-derived mesenchymal stem cells (hAMSCs).Materials and methods. For cartilage decellularization, we carried out treatment with surfactants (sodium dodecyl sulfate, Triton X-100) followed by exposure in DNAase. The metabolic activity of hAMSCs was assessed by PrestoBlue™ (Invitrogen, USA) staining. The morphological study of cell-engineered constructs (CECs) formed by culturing hAMSCs in the presence of matrices was performed using histological staining and scanning electron microscopy (SEM) with lanthanide contrasting.Results. The number of cells on the surface of both BMCH and DPAC increased within 14 days. Mitochondrial activity of the cells was 1.7, 1.7, and 1.3 times higher on days 3, 10, and 14 when cultured on DPAC compared to BMCH, respectively. On day 14 of cultivation in the chondrogenic culture medium, hAMSCs formed cell layers on the DPAC surface and on the BMCH surface. Cytoplasm of the cells included numerous granules, which, when stained, resembled the matrix itself. On the DPAC matrix surface, cells were more evenly distributed, whereas in the case of BMCH, cell adhesion and proliferation were observed only in certain areas. The ECM produced by the cells contained collagen and glycosaminoglycans (GAGs).Conclusion. The ability of DPAC obtained according to the developed protocol to form CECs with hAMSCs with uniform distribution of cells and their production of specific collagen- and GAG-containing ECM suggests that DPAC is effective in regeneration of damaged cartilage. Chondrogenic differentiation of hAMSCs was observed both when cultured with BMCH and with DPAC. When creating a tissue equivalent of cartilage in vitro, the advantage of using tissue-specific matrix over BMCH should be considered.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
49

Yoshida, Yasunori, Toshinori Takagi, Yoji Kuramoto, Kotaro Tatebayashi, Manabu Shirakawa, Kenichi Yamahara, Nobutaka Doe und Shinichi Yoshimura. „Intravenous Administration of Human Amniotic Mesenchymal Stem Cells in the Subacute Phase of Cerebral Infarction in a Mouse Model Ameliorates Neurological Disturbance by Suppressing Blood Brain Barrier Disruption and Apoptosis via Immunomodulation“. Cell Transplantation 30 (01.01.2021): 096368972110241. http://dx.doi.org/10.1177/09636897211024183.

Der volle Inhalt der Quelle
Annotation:
Neuro-inflammation plays a key role in the pathophysiology of brain infarction. Cell therapy offers a novel therapeutic option due to its effect on immunomodulatory effects. Amniotic stem cells, in particular, show promise owing to their low immunogenicity, tumorigenicity, and easy availability from amniotic membranes discarded following birth. We have successfully isolated and expanded human amniotic mesenchymal stem cells (hAMSCs). Herein, we evaluated the therapeutic effect of hAMSCs on neurological deficits after brain infarction as well as their immunomodulatory effects in a mouse model in order to understand their mechanisms of action. One day after permanent occlusion of the middle cerebral artery (MCAO), hAMSCs were intravenously administered. RT-qPCR for TNFα, iNOS, MMP2, and MMP9, immunofluorescence staining for iNOS and CD11b/c, and a TUNEL assay were performed 8 days following MCAO. An Evans Blue assay and behavioral tests were performed 2 days and several months following MCAO, respectively. The results suggest that the neurological deficits caused by cerebral infarction are improved in dose-dependent manner by the administration of hAMSCs. The mechanism appears to be through a reduction in disruption of the blood brain barrier and apoptosis in the peri-infarct region through the suppression of pro-inflammatory cytokines and the M2-to-M1 phenotype shift.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
50

Ledda, Mario, Marco Fosca, Angela De Bonis, Mariangela Curcio, Roberto Teghil, Maria Grazia Lolli, Adriana De Stefanis, Rodolfo Marchese, Julietta V. Rau und Antonella Lisi. „Placenta Derived Mesenchymal Stem Cells Hosted on RKKP Glass-Ceramic: A Tissue Engineering Strategy for Bone Regenerative Medicine Applications“. BioMed Research International 2016 (2016): 1–11. http://dx.doi.org/10.1155/2016/3657906.

Der volle Inhalt der Quelle
Annotation:
In tissue engineering protocols, the survival of transplanted stem cells is a limiting factor that could be overcome using a cell delivery matrix able to support cell proliferation and differentiation. With this aim, we studied the cell-friendly and biocompatible behavior of RKKP glass-ceramic coated Titanium (Ti) surface seeded with human amniotic mesenchymal stromal cells (hAMSCs) from placenta. The sol-gel synthesis procedure was used to prepare the RKKP glass-ceramic material, which was then deposited onto the Ti surface by Pulsed Laser Deposition method. The cell metabolic activity and proliferation rate, the cytoskeletal actin organization, and the cell cycle phase distribution in hAMSCs seeded on the RKKP coated Ti surface revealed no significant differences when compared to the cells grown on the treated plastic Petri dish. The health of of hAMSCs was also analysed studying the mRNA expressions of MSC key genes and the osteogenic commitment capability using qRT-PCR analysis which resulted in being unchanged in both substrates. In this study, the combination of the hAMSCs’ properties together with the bioactive characteristics of RKKP glass-ceramics was investigated and the results obtained indicate its possible use as a new and interesting cell delivery system for bone tissue engineering and regenerative medicine applications.
APA, Harvard, Vancouver, ISO und andere Zitierweisen
Wir bieten Rabatte auf alle Premium-Pläne für Autoren, deren Werke in thematische Literatursammlungen aufgenommen wurden. Kontaktieren Sie uns, um einen einzigartigen Promo-Code zu erhalten!

Zur Bibliographie