28 results on '"human adipose tissue–derived mesenchymal stem cells"'
Search Results
2. INVESTIGATION OF URSODEOXYCHOLIC ACID EFFECTS ON SIROLIMUS TREATED ADIPOSE TISSUE-DERIVED MESENCHYMAL STEM CELLS.
- Author
-
ARISU NAGHAVI, Esra, GOKER BAGCA, Bakiye, TEKELI, Senem, YIGITTURK, Gurkan, GOKCE, Burak, TOMRUK, Canberk, CAVUSOGLU, Turker, BIRAY AVCI, Cigir, GUNDUZ, Cumhur, and UYANIKGIL, Yigit
- Subjects
- *
URSODEOXYCHOLIC acid , *RAPAMYCIN , *ADIPOSE tissues , *MESENCHYMAL stem cells , *IMMUNOSUPPRESSIVE agents - Abstract
Objective: The usage of mesenchymal stem cells (MSC) with immunosuppressive drugs after organ transplantation is becoming remarkable in clinical applications. However, the drugs negatively affect MSCs. Ursodeoxycholic acid (UDCA), which is an antioxidant molecule, may reverse these effects. The study aims that to determine the effects of sirolimus and UDCA on human adipose tissue-derived MSCs (ADMSCs) individually and in combination. Material and Method: The cytotoxicity of the agents was evaluated by WST-1 test in time and dose-dependent manner. The combinational effects were determined using isobologram analysis. Muse cell analyzer was used for the evaluation of apoptosis and cell cycle. Oxidative stress markers were measured by biochemical methods. Results: IC50 dose of sirolimus was determined as 18.58µM in the 48th hour. Because no cytotoxic effect was observed at the studied doses of UDCA, the apoptosis, cell cycle, and oxidative stress indicator analyses were continued with a safe dose of 100 µM. Sirolimus promoted apoptosis and inhibited cell proliferation. It was determined that UDCA reduced the apoptotic and anti-proliferative effects of sirolimus on ADMSCs with its anti-oxidant property. Conclusion: The UDCA treatment in combination with immunosuppressive therapy after organ and tissue transplantation may have positive effects on ADMSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
3. First identification of the effects of low frequency electromagnetic field on the micromolecular changes in adipose tissue-derived mesenchymal stem cells by fourier transform infrared spectroscopy
- Author
-
Kornelia Łach, Józef Cebulski, Radosław Chaber, Beata Kocan, Renata Wojnarowska-Nowak, and Agnieszka Banaś-Ząbczyk
- Subjects
adipose tissue-mesenchymal stem cells ,electromagnetic field ,fourier transform infrared spectroscopy ,human adipose tissue-derived mesenchymal stem cells ,mesenchymal stem cell ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Purpose: In this study, we hypothesize that exposure of adipose tissue-mesenchymal stem cells (AT-MSCs) to electromagnetic field (EMF) may impact adipose stem cells' micromolecular structure (analyzed using Fourier transform infrared spectroscopy [FTIR]). Materials and Methods: The AT-MSCs were exposed to continuous vertically applied sinusoidal EMF with a frequency of 50 Hz and a flux density of 1.5 mT for 24, 48, and 72 h. After an appropriate time (24, 48, 72 h) cells were washed with PBS, scrubbed, and immediately taken into FTIR analyses. Results: EMFs affect AT-MSCs. The greatest differences were in the range of nucleic acids and proteins in the fingerprint region which occurred after 24 and 48 h of EMF exposure. However, in the case of 72 h of EMF exposure, no significant differences were noticed in the FTIR spectra towards the control. Conclusions: FTIR spectra show differences between samples under the influence of EMF before they will be manifested at the morphological level. The largest differences in the range of nucleic acids and proteins in the fingerprint region occurred at 24 and 48 h of EMF exposure. That means it was during the first 48 h after EMF exposure a great number of dynamic changes occurred. However, in the case of AT-MSCs in 72 h EMF and 72 h control, no significant differences were noted in the FTIR spectra, which means that the chemical composition in these two cases is similar. EMF is not neutral for stem cells, especially in the in the first hours of interaction (24 h, 48 h).
- Published
- 2021
- Full Text
- View/download PDF
4. Reversibility of hAT-MSCs phenotypic and metabolic changes after exposure to and withdrawal from HCC-conditioned medium through regulation of the ROS/MAPK/HIF-1α signaling pathway
- Author
-
Chenyang Wang, Jie Hu, Zheng Chen, Yifan Wang, Sinan Lu, Yuan Zhang, Yufeng Li, Yucheng Xiang, Yutian Ji, Cheng Zeng, Yuan Ding, and Weilin Wang
- Subjects
Hepatocellular carcinoma ,Conditioned medium ,Human adipose tissue-derived mesenchymal stem cells ,Cell phenotype ,Glucose metabolism ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Mesenchymal stem cells (MSCs) play an important role in tumor progression; concomitantly, MSCs also undergo profound changes in the tumor microenvironment (TME). These changes can directly impact the application and efficacy of MSC-based anti-tumor therapy. However, few studies have focused on the regulation of MSC fate in TME, which will limit the progress of MSC-based anti-tumor therapy. Herein, we investigated the effects of conditioned medium from human hepatocellular carcinoma cells (HCC-CM) on the phenotype and glucose metabolism of human adipose tissue-derived MSCs (hAT-MSCs). Methods The passage 2 (P2) to passage 3 (P3) hAT-MSCs were exposed to conditioned medium from Hep3B, Huh7 and HCCLM3 cells for 4–8 weeks in vitro. Then, immunofluorescent, CCK-8 assay, EdU assay, Transwell assay, and flow cytometry were used to assess the alterations in cell phenotype in terms of cell morphology, secretory profiles, proliferation, migration, invasion, cell cycle, and apoptosis. In addition, glucose metabolism was evaluated by related kits. Next, the treated hAT-MSCs were subjected to withdrawal from HCC-CM for 2–4 weeks, and alterations in phenotype and glucose metabolism were reevaluated. Finally, the molecular mechanism was clarified by Western blotting. Results The results revealed that after exposure to HCC-CM, hAT-MSCs developed a stellate-shaped morphology. In association with cytoskeleton remodeling, hAT-MSCs showed enhanced capacities for migration and invasion, while cell proliferation was inhibited by regulating the cell cycle by downregulating cyclins and cyclin-dependent kinases and activating the mitochondrial apoptosis pathway. In terms of glucose metabolism, our results showed mitochondrial dysfunction and elevated glycolysis of hAT-MSCs. However, interestingly, when the treated hAT-MSCs were subjected to withdrawal from HCC-CM, the alterations in phenotype and glucose metabolism could be reversed, but secretory phenotype and tumor-promoting properties appear to be permanent. Further studies showed that these changes in hAT-MSCs may be regulated by the ROS/MAPK/HIF-1α signaling pathway. Conclusion Taken together, the effects of long-term HCC-CM treatment on phenotype and glucose metabolism in hAT-MSCs are modest and largely reversible after withdrawal, but HCC-CM endow hAT-MSCs with permanent secretory phenotype and tumor-promoting properties. This is the first report on the reversal of phenotype and glucose metabolism in tumor-associated MSCs (TA-MSCs), it is anticipated that new insights into TA-MSCs will lead to the development of novel strategies for MSC-based anti-tumor therapy.
- Published
- 2020
- Full Text
- View/download PDF
5. First Identification of the Effects of Low Frequency Electromagnetic Field on the Micromolecular Changes in Adipose Tissue-Derived Mesenchymal Stem Cells by Fourier Transform Infrared Spectroscopy.
- Author
-
Łach, Kornelia, Cebulski, Józef, Chaber, Radosław, Kocan, Beata, Wojnarowska-Nowak, Renata, and Banaś-Ząbczyk, Agnieszka
- Subjects
FOURIER transform infrared spectroscopy ,MESENCHYMAL stem cells ,ELECTROMAGNETIC fields ,ADIPOSE tissues ,PEPTIDE mass fingerprinting ,FAT cells ,NEAR infrared spectroscopy - Abstract
Purpose: In this study, we hypothesize that exposure of adipose tissue-mesenchymal stem cells (AT-MSCs) to electromagnetic field (EMF) may impact adipose stem cells' micromolecular structure (analyzed using Fourier transform infrared spectroscopy [FTIR]). Materials and Methods: The AT-MSCs were exposed to continuous vertically applied sinusoidal EMF with a frequency of 50 Hz and a flux density of 1.5 mT for 24, 48, and 72 h. After an appropriate time (24, 48, 72 h) cells were washed with PBS, scrubbed, and immediately taken into FTIR analyses. Results: EMFs affect AT-MSCs. The greatest differences were in the range of nucleic acids and proteins in the fingerprint region which occurred after 24 and 48 h of EMF exposure. However, in the case of 72 h of EMF exposure, no significant differences were noticed in the FTIR spectra towards the control. Conclusions: FTIR spectra show differences between samples under the influence of EMF before they will be manifested at the morphological level. The largest differences in the range of nucleic acids and proteins in the fingerprint region occurred at 24 and 48 h of EMF exposure. That means it was during the first 48 h after EMF exposure a great number of dynamic changes occurred. However, in the case of AT-MSCs in 72 h EMF and 72 h control, no significant differences were noted in the FTIR spectra, which means that the chemical composition in these two cases is similar. EMF is not neutral for stem cells, especially in the in the first hours of interaction (24 h, 48 h). [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
6. H19/miR‐30a/C8orf4 axis modulates the adipogenic differentiation process in human adipose tissue‐derived mesenchymal stem cells.
- Author
-
Li, Kun, Wu, Yue, Yang, Hao, Hong, Pengyu, Fang, Xiaodan, and Hu, Yanjia
- Subjects
- *
ADIPOGENESIS , *MESENCHYMAL stem cells , *NON-coding RNA , *MESSENGER RNA , *PROTEIN expression , *PROTEIN binding - Abstract
The adipogenic differentiation of adipose tissue‐derived mesenchymal stem cells (ADSCs) is a critical issue in many obesity‐related disorders. Cytidine‐cytidine‐adenosine‐adenosine‐thymidine (CCAAT) enhancer binding protein α (CEBP‐α) and peroxisome proliferator‐activated receptor‐γ are two important lipogenic and adipogenic transcription factors and markers in adipogenic differentiation. Noncoding RNAs participate in adipogenic differentiation. The long noncoding RNA (lncRNA) H19 is related to multiple cellular differentiation, including adipogenic differentiation; however, its function and precise molecular mechanism in human ADSCs (hADSCs) adipogenic differentiation are unclear. microRNAs that were differentially expressed in adipogenic differentiation and could be targeted by H19 were screened and selected; the regulation and interaction between H19 and miR‐30a were verified. The interaction between miR‐30a and predicted downstream target C8orf4 was validated. The dynamic effects of H19 and miR‐30a on C8orf4 messenger RNA (mRNA) expression and protein and adipogenic differentiation were evaluated. miR‐30a negatively regulated H19 with each other through direct binding. As predicted by TargetScan and verified using luciferase reporter gene assays, miR‐30a directly bound to the 3′‐untranslated region of C8orf4 to inhibit its expression; H19 knockdown suppressed while miR‐30a inhibition promoted the mRNA expression and the protein levels of C8orf4 and adipogenic differentiation; the effect of H19 knockdown could be partially reversed by miR‐30a inhibition. The lncRNA H19 serves as a competing endogenous RNA (ceRNA) for miR‐30a to augment miR‐30a downstream target C8orf4, therefore modulating adipogenic differentiation in hADSCs. From the perspective of lncRNA–miRNA–mRNA regulation, we provided a novel regulatory mechanism of hADSCs adipogenic differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
7. β-Lapachone Regulates Obesity through Modulating Thermogenesis in Brown Adipose Tissue and Adipocytes: Role of AMPK Signaling Pathway.
- Author
-
Kwak, Hyun Jeong, Jeong, Mi-Young, Um, Jae-Young, and Park, Jinbong
- Subjects
- *
ADIPOSE tissues , *ANIMAL experimentation , *BARK , *BODY temperature regulation , *CELL culture , *CELL differentiation , *CELL separation , *DRUG design , *CLINICAL drug trials , *FAT cells , *MICE , *MITOCHONDRIA , *MOLECULAR structure , *OBESITY , *PHOSPHATASES , *POLYMERASE chain reaction , *RESEARCH funding , *STAINS & staining (Microscopy) , *STEM cells , *WESTERN immunoblotting , *PHYTOCHEMICALS , *REVERSE transcriptase polymerase chain reaction , *DATA analysis software , *CELL survival , *SIGNAL peptides , *MANN Whitney U Test , *KRUSKAL-Wallis Test , *IN vivo studies - Abstract
Activation of brown adipose tissue (BAT) has been proposed as a promising target against obesity due to its increased capacity for thermogenesis. In this study, we explored the effect of β -Lapachone (β L), a compound obtained from the bark of the lapacho tree, against obesity. In vivo administration of β L into either high fat diet (HFD)-induced obese C57BL6 mice and genetically obese Lepr - ∕ - mice prevented body weight gain, which was associated with tissue weight loss of white adipose tissue (WAT). In addition, β L elevated thermogenic proteins including uncoupling protein 1 (UCP1) and mitochondrial count in BAT and human adipose tissue-derived mesenchymal stem cells (hAMSCs). β L also induced AMP-activated protein kinase (AMPK) phosphorylation, subsequent upregulation of acetyl-CoA carboxylase (ACC) and UCP1, and these effects were diminished by AMPK inhibitor compound C, suggesting that AMPK underlies the effects of β L. Mitogen-activated protein kinase pathways participated in the thermogenesis of β L, specifically p38, c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase 1/2 (ERK1/2) were activated by β L treatment in hAMSCs. Additionally, inhibitors of p38/JNK/ERK1/2 abrogated the activity of β L. Taken together, β L exerts anti-obese effects by inducing thermogenesis mediated by AMPK signaling pathway, suggesting that β L may have a potential therapeutic implication of obesity. Taken together, β L exerts anti-obese effects by not only inducing thermogenesis on brown adipocytes but also inducing the browning of white adipocytes. The anti-obese effect of β L is mediated by AMPK signaling pathway, suggesting that β L may have potential therapeutic implication of obesity. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
8. Kojyl cinnamate esters are peroxisome proliferator-activated receptor α/γ dual agonists.
- Author
-
Kim, Sae On, Han, Yujia, Ahn, Sungjin, An, Seungchan, Shin, Jeayoung C., Choi, Hyunjung, Kim, Hyoung-June, Park, Nok Hyun, Kim, Yong-Jin, Jin, Sun Hee, Rho, Ho Sik, and Noh, Minsoo
- Subjects
- *
ADIPOSE tissues , *MESENCHYMAL stem cells , *ADIPONECTIN , *PEROXISOME proliferator-activated receptors , *ADIPOGENESIS - Abstract
Graphical abstract Abstract Adiponectin is an adipocytokine with insulin-sensitizing, anti-inflammatory, anti-atherosclerotic, and anti-aging properties. Compounds with the ability to promote adiponectin secretion are of interest for the development of anti-aging drugs to improve skin-aging phenotypes. In the phenotypic assay to measure adiponectin secretion during adipogenesis in human adipose tissue-derived mesenchymal stem cells (hAT-MSCs), kojyl cinnamate ester derivatives increased adiponectin secretion. A target identification study showed that the kojyl cinnamate ester derivatives competitively bound to peroxisome proliferator-activated receptor α/γ (PPARα/γ). The upregulation of adiponectin production induced by kojyl cinnamate ester derivatives was significantly correlated with PPARα and PPARγ binding activities. Kojyl cinnamate ester derivatives significantly increased the transcription of genes encoding cholesterol and fatty acid synthesizing enzymes in hAT-MSCs. Notably, the kojyl cinnamate esters upregulated the gene transcription of lipid metabolic enzymes in human epidermal keratinocytes, which are important in the integrity of skin permeability barrier. In addition, the kojyl cinnamate esters that function as PPARα/γ dual modulators inhibited ultraviolet B irradiation-induced inflammation in human epidermal keratinocytes. Therefore, kojyl cinnamate ester derivatives are a novel class of PPARα/γ dual agonists with the potential to improve skin-aging phenotypes. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
9. INVESTIGATION OF URSODEOXYCHOLIC ACID EFFECTS ON SIROLIMUS TREATED ADIPOSE TISSUE-DERIVED MESENCHYMAL STEM CELLS
- Author
-
Çavuşoğlu, Türker, Arisu, Esra, Bağca, Bakiye Göker, None, Senem, Yiğittürk, Gürkan, Gökçe, Burak, and Tomruk, Canberk
- Subjects
Sirolimus ,Ursodeoxycholic acid ,Oxidative stress ,Human adipose tissue-derived mesenchymal stem cells - Abstract
Objective The usage of mesenchymal stem cells (MSC) with immunosuppressive drugs after organ transplantation is becoming remarkable in clinical applications. However, the drugs negatively affect MSCs. Ursodeoxycholic acid (UDCA), which is an antioxidant molecule, may reverse these effects. The study aims that to determine the effects of sirolimus and UDCA on human adipose tissue-derived MSCs (ADMSCs) individually and in combination. Material and Method The cytotoxicity of the agents was evaluated by WST-1 test in time and dose-dependent manner. The combinational effects were determined using isobologram analysis. Muse cell analyzer was used for the evaluation of apoptosis and cell cycle. Oxidative stress markers were measured by biochemical methods. Results IC50 dose of sirolimus was determined as 18.58?M in the 48th hour. Because no cytotoxic effect was observed at the studied doses of UDCA, the apoptosis, cell cycle, and oxidative stress indicator analyses were continued with a safe dose of 100 ?M. Sirolimus promoted apoptosis and inhibited cell proliferation. It was determined that UDCA reduced the apoptotic and anti-proliferative effects of sirolimus on ADMSCs with its anti-oxidant property. Conclusion The UDCA treatment in combination with immunosuppressive therapy after organ and tissue transplantation may have positive effects on ADMSCs.
- Published
- 2022
10. Obesity-associated miR-148a is regulated by cytokines and adipokines via a transcriptional mechanism.
- Author
-
CHUNMEI SHI, LINGXIA PANG, CHENBO JI, JIAQIN WANG, NING LIN, JIANTAO CHEN, LING CHEN, LEI YANG, FANGYAN HUANG, YAHUI ZHOU, XIRONG GUO, HUI LIANG, and MIN ZHANG
- Subjects
- *
MICRORNA , *OBESITY genetics , *CYTOKINES , *ADIPOKINES , *GENE regulatory networks , *MESENCHYMAL stem cells , *ADIPOSE tissues , *GENE expression - Abstract
Our previous study revealed that miR-148a, a cyclic adenosine monophosphate-response element binding protein-modulated microRNA that promotes adipocyte differentiation by inhibiting Wnt1, is a biomarker of obesity in human subjects and a mouse model. The present study investigated the expression of miR-148a in human adipose tissue-derived mesenchymal stem cells (hMSCs-Ad) in response to inflammatory cytokines and adipokines to clarify its underlying mechanism. miR-148a expression was detected using reverse transcription-quantitative polymerase chain reaction analysis and its promoter activity was detected with a luciferase assay. miR-148a expression levels decreased when differentiated hMSCs-Ad were exposed to inflammatory cytokines or adipokines, which suggested that miR-148a may be important in adipocyte metabolism and inflammation. Furthermore, the promoter activity of miR-148a decreased following treatment of cells with inflammatory cytokines or adipokines. The results of the present study indicated a novel role of miR-148a in adipocyte inflammation; therefore, miR-148a may be involved in obesity complications via its own underlying transcriptional mechanism. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
11. Vanillin attenuates negative effects of ultraviolet A on the stemness of human adipose tissue-derived mesenchymal stem cells.
- Author
-
Lee, Sang Yeol, Park, See-Hyoung, Kim, Mi Ok, Lim, Inhwan, Kang, Mingyeong, Oh, Sae Woong, Jung, Kwangseon, Jo, Dong Gyu, Cho, Il-Hoon, and Lee, Jongsung
- Subjects
- *
VANILLIN , *DRUG side effects , *ULTRAVIOLET radiation , *ADIPOSE tissues , *MESENCHYMAL stem cells - Abstract
Ultraviolet A (UVA) irradiation induces various changes in cell biology. The objective of this study was to determine the effect of vanillin on UVA irradiation-induced damages in the stemness properties of human adipose tissue-derived mesenchymal stem cells (hAMSCs). UVA-antagonizing mechanisms of vanillin were also examined. The results revealed that vanillin attenuated UVA-induced reduction of the proliferative potential and stemness of hAMSCs evidenced by increased proliferative activity in BrdU incorporation assay and upregulation of stemness-related genes (OCT4, NANOG and SOX2) in response to vanillin treatment. UVA-induced reduction in mRNA level of hypoxia-inducible factor (HIF)-1α was significantly recovered by vanillin. In addition, the antagonizing effect of vanillin on UVA was found to be mediated by reduced production of PGE 2 through inhibiting JNK and p38 MAPK. Taken together, these findings showed that vanillin could improve the reduced stemness of hAMSCs induced by UVA. The effect of vanillin is mediated by upregulating HIF-1α via inhibiting PGE 2 -cAMP signaling. Therefore, vanillin might be used as an antagonizing agent to mitigate the effects of UVA. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
12. Simvastatin coating of TiO2 scaffold induces osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells.
- Author
-
Pullisaar, Helen, Reseland, Janne E., Haugen, Håvard J., Brinchmann, Jan E., and Østrup, Esben
- Subjects
- *
SIMVASTATIN , *TITANIUM dioxide , *ADIPOSE tissues , *MESENCHYMAL stem cells , *TISSUE engineering , *ALKALINE phosphatase - Abstract
Highlights: [•] Tissue engineering approach combining MSCs, simvastatin and a 3D porous scaffold. [•] Induction of osteogenic differentiation in MSCs by simvastatin coated scaffolds. [•] Lower concentration of simvastatin needed in 3D culture than reported for 2D culture. [•] A strategy for human adipose tissue-derived MSC based bone tissue engineering. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
13. Reversibility of hAT-MSCs phenotypic and metabolic changes after exposure to and withdrawal from HCC-conditioned medium through regulation of the ROS/MAPK/HIF-1α signaling pathway
- Author
-
Yucheng Xiang, Yuan Ding, Chenyang Wang, Zheng Chen, Cheng Zeng, Jie Hu, Yutian Ji, Yuan Zhang, Yifan Wang, Sinan Lu, Yu-Feng Li, and Wei-Lin Wang
- Subjects
MAPK/ERK pathway ,Carcinoma, Hepatocellular ,Hepatocellular carcinoma ,Medicine (miscellaneous) ,Cell morphology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,lcsh:Biochemistry ,Tumor Microenvironment ,Humans ,lcsh:QD415-436 ,Conditioned medium ,Cell Proliferation ,lcsh:R5-920 ,Cell phenotype ,Glucose metabolism ,Kinase ,Cell growth ,Chemistry ,Research ,Mesenchymal stem cell ,Liver Neoplasms ,Mesenchymal Stem Cells ,Cell Biology ,Cell cycle ,Cell biology ,Phenotype ,Culture Media, Conditioned ,Molecular Medicine ,Stem cell ,Signal transduction ,Human adipose tissue-derived mesenchymal stem cells ,lcsh:Medicine (General) ,Reactive Oxygen Species ,Signal Transduction - Abstract
Background Mesenchymal stem cells (MSCs) play an important role in tumor progression; concomitantly, MSCs also undergo profound changes in the tumor microenvironment (TME). These changes can directly impact the application and efficacy of MSC-based anti-tumor therapy. However, few studies have focused on the regulation of MSC fate in TME, which will limit the progress of MSC-based anti-tumor therapy. Herein, we investigated the effects of conditioned medium from human hepatocellular carcinoma cells (HCC-CM) on the phenotype and glucose metabolism of human adipose tissue-derived MSCs (hAT-MSCs). Methods The passage 2 (P2) to passage 3 (P3) hAT-MSCs were exposed to conditioned medium from Hep3B, Huh7 and HCCLM3 cells for 4–8 weeks in vitro. Then, immunofluorescent, CCK-8 assay, EdU assay, Transwell assay, and flow cytometry were used to assess the alterations in cell phenotype in terms of cell morphology, secretory profiles, proliferation, migration, invasion, cell cycle, and apoptosis. In addition, glucose metabolism was evaluated by related kits. Next, the treated hAT-MSCs were subjected to withdrawal from HCC-CM for 2–4 weeks, and alterations in phenotype and glucose metabolism were reevaluated. Finally, the molecular mechanism was clarified by Western blotting. Results The results revealed that after exposure to HCC-CM, hAT-MSCs developed a stellate-shaped morphology. In association with cytoskeleton remodeling, hAT-MSCs showed enhanced capacities for migration and invasion, while cell proliferation was inhibited by regulating the cell cycle by downregulating cyclins and cyclin-dependent kinases and activating the mitochondrial apoptosis pathway. In terms of glucose metabolism, our results showed mitochondrial dysfunction and elevated glycolysis of hAT-MSCs. However, interestingly, when the treated hAT-MSCs were subjected to withdrawal from HCC-CM, the alterations in phenotype and glucose metabolism could be reversed, but secretory phenotype and tumor-promoting properties appear to be permanent. Further studies showed that these changes in hAT-MSCs may be regulated by the ROS/MAPK/HIF-1α signaling pathway. Conclusion Taken together, the effects of long-term HCC-CM treatment on phenotype and glucose metabolism in hAT-MSCs are modest and largely reversible after withdrawal, but HCC-CM endow hAT-MSCs with permanent secretory phenotype and tumor-promoting properties. This is the first report on the reversal of phenotype and glucose metabolism in tumor-associated MSCs (TA-MSCs), it is anticipated that new insights into TA-MSCs will lead to the development of novel strategies for MSC-based anti-tumor therapy.
- Published
- 2020
14. PPARγ silencing enhances osteogenic differentiation of human adipose-derived mesenchymal stem cells.
- Author
-
Lee, Mon‐Juan, Chen, Hui‐Ting, Ho, Mei‐Ling, Chen, Chung‐Hwan, Chuang, Shu‐Chun, Huang, Sung‐Cheng, Fu, Yin‐Chih, Wang, Gwo‐Jaw, Kang, Lin, and Chang, Je‐Ken
- Subjects
PEROXISOME proliferator-activated receptors ,GENE silencing ,BONE morphogenetic proteins ,ADIPOSE tissues ,MESENCHYMAL stem cells ,CELL differentiation ,ADIPOGENESIS - Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is the master regulator of adipogenesis, and has been indicated as a potential therapeutic target to promote osteoblast differentiation. However, recent studies suggest that suppression of PPARγ inhibits adipogenesis, but does not promote osteogenic differentiation in human bone marrow-derived mesenchymal stem cells ( hBMSCs). It was reasoned that the osteogenic effect of PPARγ suppression may be masked by the strong osteogenesis-inducing condition commonly used, resulting in a high degree of matrix mineralization in both control and experimental groups. This study investigates the role of PPARγ in the lineage commitment of human adipose-derived mesenchymal stem cells ( hADSCs) by interfering with the function of PPARγ mRNA through small interfering RNAs (si RNAs) specific for PPARγ2. By applying an osteogenic induction condition less potent than that used conventionally, we found that PPARγ silencing led to retardation of adipogenesis and stimulated a higher level of matrix mineralization. The mRNA level of PPARγ decreased to 47% of control 2 days after treatment with 50 nmol/l PPARγ2 si RNA, while its protein expression was 60% of mock control. In the meantime, osteogenic marker genes, including bone morphogenic protein 2 ( BMP2), runt-related transcription factor 2 (Runx2), alkaline phosphatase ( ALP) and osteocalcin ( OC), were up-regulated under PPARγ silencing. Our results suggest that transient suppression of PPARγ promotes the onset of osteogenesis, and may be considered a new strategy to stimulate bone formation in bone tissue engineering using hADSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
15. Functional expression of smooth muscle-specific ion channels in TGF-β1-treated human adipose-derived mesenchymal stem cells.
- Author
-
Won Sun Park, Soon Chul Heo, Eun Su Jeon, Da Hye Hong, Youn Kyoung Son, Jae-Hong Ko, Hyoung Kyu Kim, Sun Young Lee, Jae Ho Kim, and Jin Han
- Subjects
- *
VASCULAR smooth muscle , *ION channels , *TRANSFORMING growth factors , *MESENCHYMAL stem cells , *GENE expression , *ADIPOSE tissues , *CELL differentiation , *CARTILAGE cells - Abstract
Human adipose tissue-derived mesenchymal stem cells (hASCs) have the power to differentiate into various cell types including chondrocytes, osteocytes, adipocytes, neurons, cardiomyocytes, and smooth muscle cells. We characterized the functional expression of ion channels after transforming growth factor-β1 (TGF-β1)-induced differentiation of hASCs, providing insights into the differentiation of vascular smooth muscle cells. The treatment of hASCs with TGF-β1 dramatically increased the contraction of a collagen-gel lattice and the expression levels of specific genes for smooth muscle including α-smooth muscle actin, calponin, smooth mucle-myosin heavy chain, smoothelin-B, myocardin, and h-caldesmon. We observed Ca2+, big-conductance Ca2+-activated K+ (BKCa), and voltage-dependent K+ (Kv) currents in TGF-β1-induced, differentiated hASCs and not in undifferentiated hASCs. The currents share the characteristics of vascular smooth muscle cells (SMCs). RT-PCR and Western blotting revealed that the L-type (Cav1.2) and T-type (Cav3.1, 3.2, and 3.3), known to be expressed in vascular SMCs, dramatically increased along with the Cavβ1 and Cavβ3 subtypes in TGF-β1-induced, differentiated hASCs. Although the expression-level changes of the β-subtype BKCa channels varied, the major α-subtype BKCa channel (KCa1.1) clearly increased in the TGF-β1-induced, differentiated hASCs. Most of the Kv subtypes, also known to be expressed in vascular SMCs, dramatically increased in the TGF-β1-induced, differentiated hASCs. Our results suggest that TGF-β1 induces the increased expression of vascular SMC-like ion channels and the differentiation of hASCs into contractile vascular SMCs. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
16. Efficacy and safety of human adipose tissue-derived mesenchymal stem cells for supporting hematopoiesis.
- Author
-
Nishiwaki, Satoshi, Nakayama, Takayuki, Saito, Shigeki, Mizuno, Hiroki, Ozaki, Takenori, Takahashi, Yoshiyuki, Maruyama, Shoichi, Nishida, Tetsuya, Murata, Makoto, Kojima, Seiji, and Naoe, Tomoki
- Abstract
We have demonstrated that adipose tissue-derived mesenchymal stem cells (ADSCs) from mice are capable of reconstituting the hematopoietic microenvironment, and facilitate hematopoiesis more effectively than bone marrow-derived mesenchymal stem cells (BMSCs) in mouse. The ready accessibility of fat tissue rich in MSCs and the higher hematopoiesis-supporting capacities of ADSCs suggest that ADSCs might represent a new therapeutic modality for the regeneration of impaired hematopoiesis. As a further step towards their use in clinical practice, we established human BMSCs and ADSCs from healthy volunteers of similar age, and compared their proliferation capacities, hematopoiesis-supporting properties, and safety. In vitro cell proliferation studies revealed that ADSCs have a higher population doubling number than BMSCs. In vitro co-culture assays showed that ADSCs not only support human CD34 peripheral blood stem cells (PBSCs), but also yield significantly more non-adherent hematic cells than BMSCs. In vitro progenitor assays revealed that ADSCs promote a higher frequency of early progenitors than do BMSCs. Interestingly, BM cellularity in irradiated mice that had received ADSCs tended to be higher than that of mice treated with BMSCs. When MSCs were injected into the BM cavity of tibiae, we observed no evidence of MSC-induced toxicity either during or after treatment. In addition, no microscopic abnormalities were observed in the bone marrow and major organs. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
17. Insulin-producing cells from human adipose tissue-derived mesenchymal stem cells detected by atomic force microscope.
- Author
-
Shi, Qiping, Luo, Simin, Jin, Hua, Cai, Jiye, Jia, Haiying, Feng, Lie, and Lu, Xiaohua
- Subjects
- *
ADIPOSE tissues , *MESENCHYMAL stem cells , *INSULIN biotechnology , *ATOMIC force microscopy , *GLUCOSE synthesis - Abstract
We successfully differentiated human adipose tissue-derived mesenchymal stem cells (haMSCs) into insulin-producing cells (IPCs) in vitro and did not use any insulin which might be absorbed by cells during in vitro culture. Expression of insulin gene was massively increased by 28,000-fold at day 12 compared with haMSCs ( P < 0.05). IPCs could secrete insulin after glucose was stimulated. The higher the concentration of glucose, the more production of insulin was noted. We reported AFM images of IPCs for the first time. AFM images showed that the sizes of cells were similar to each other, and all IPC surface had a porous structure in the cytoplasm area. In sugar-free group, the size of holes was similar (diameter, 1,086.98 ± 156.70 nm; depth, 185.22 ± 52.14 nm). In higher sugar-stimulated group, there were more holes with bigger diameter and smaller depth. (diameter, 3,183.65 ± 2,229.18 nm; depth 109.42 ± 56.26 nm, P < 0.05). We found that the hole diameter and depth could change with the concentration of glucose in media. Concurrently, laser scanning confocal microscopy images indicated that cortical actin network beneath plasma membrane in IPCs was dense and continuous. After glucose stimulation, we found the actin web depolymerized and became discontinuous in IPCs. We speculated that diameter augmentation of holes located in the cytoplasm area in IPCs was one manifestation of excytosis increase. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
18. MiR-21 Regulates Adipogenic Differentiation through the Modulation of TGF-β Signaling in Mesenchymal Stem Cells Derived from Human Adipose Tissue.
- Author
-
Yeon Jeong Kim, Soo Jin Hwang, Yong Chan Bae, and Jin Sup Jung
- Subjects
ADIPOSE tissue transplantation ,TRANSFORMING growth factors ,CELLULAR therapy ,METABOLIC disorders ,CELL differentiation ,CHEMICAL reactions - Abstract
A better understanding of the molecular mechanisms that govern human adipose tissue-derived mesenchymal stem cells (hASCs) differentiation could improve hASCs-based cell therapy and provide new insights into a number of diseases, including obesity. In this study, we examined the roles of microRNA-21 (miR-21) in adipogenic differentiation of hASCs. We found that miR-21 expression was transiently increased after induction of adipogenic differentiation, peaked at 3 days, and returned to the baseline level 8 days. Lentiviral overexpression of miR-21 enhanced adipogenic differentiation. Overexpression of miR-21 decreased both protein and mRNA levels of TGFBR2. The expression of TGFBR2 was decreased during adipogenic differentiation of hASCs in concordance with an increase in the level of miR-21. In contrast, inhibiting miR-21 with 2′-O-methyl-antisense microRNA increased TGFBR2 protein levels in hASCs, accompanied by decreased adipogenic differentiation. The activity of a luciferase construct containing the miR-21 target site from the TGFBR2 3′UTR was lower in LV-miR21-infected hASCs than in LV-miLacZ infected cells. TGF-β-induced inhibition of adipogenic differentiation was significantly decreased in miR-21 overexpressing cells compared with control lentivirus-transduced cells. RNA interference-mediated downregulation of SMAD3, but not of SMAD2, increased adipogenic differentiation. Overexpression and inhibition of miR-21 altered SMAD3 phosphorylation without affecting total levels of SMAD3 protein. Our data are the first to demonstrate that the role of miR-21 in the adipogenic differentiation of hASCs is mediated through the modulation of TGF-βsignaling. This study improves our knowledge of the molecular mechanisms governing hASCs differentiation, which may underlie the development of obesity or other metabolic diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
19. miR-196a Regulates Proliferation and Osteogenic Differentiation in Mesenchymal Stem Cells Derived From Human Adipose Tissue.
- Author
-
Yeon Jeong Kim, Sang Woo Bae, Sung Sook Yu, Yong Chan Bae, and Jin Sup Jung
- Abstract
The article presents a study investigating the role of microRNA (miRNA)-196a on the proliferation of human adipose tissue-derived mesenchymal stem cells (hASCs) and osteogenic differentiation. The observation that lentiviral overexpression of miR-196a decreased the proliferation of hASC and supported osteogenic differentiation is noted, mentioning that adipogenic differentiation was not affected.
- Published
- 2009
- Full Text
- View/download PDF
20. Ursodeoksikolik asidin sirolimusa maruz kalmış yağ doku kaynaklı mezenkimal kök hücre hattı üzerine etkilerinin incelenmesi
- Author
-
Arısu Naghavi, Esra, Uyanıkgil, Yiğit, Kök Hücre Anabilim Dalı, and Sağlık Bilimleri Enstitüsü
- Subjects
Sirolimus ,Oksidatif Stres ,Oxidative Stress ,Yağ Doku Kaynaklı Mezenkimal Kök Hücre ,Ursodeoxycholic Acid ,Ursodeoksikolik Asit ,Human Adipose Tissue-Derived Mesenchymal Stem Cells ,Biology ,Biyoloji - Abstract
Mezenkimal kök hücre (MKH) tedavileri günümüzde tıbbın birçok alanında deneysel ve klinik olarak uygulamaya girmiş önemli tedavi yöntemleridir. Klinik vakalarda organ nakli sonrasında immunsupresif ilaçların yanında destek amaçlı olarak hastalara MKH nakledilmesi gittikçe yaygınlaşmaktadır. Yapılan bu çalışmadaki amaç organ nakillerinde immunsupresif amaçla kullanılan Sirolimus’un, insan yağ doku kaynaklı mezenkimal kök hücreler üzerindeki sitotoksik etkilerinin araştırılması ve antioksidan özelliği bilinen Ursodeoksikolik asitin (UDKA), bu toksisite üzerine etkilerinin araştırılmasıdır. İnsan yağ doku kaynaklı mezenkimal kök hücre hattı ticari olarak temin edilmiştir. Özel besiyerleri içinde üretilen insan mezenkimal kök hücreleri yeterli sayıya ulaşana kadar 37 ° C’de inkübe edilmiştir. Etken maddelerin sitotoksik etkileri WST-1 testi ile zaman ve doz bağımlı olarak değerlendirilmiştir. Canlılık ve proliferasyon analizleri için ışık mikroskobu ve hücre sayım cihazı, apoptoz ve hücre siklusu değerlendirmeleri için ise Muse® hücre analiz cihazı kullanılmıştır. Biyokimyasal yöntemlerle lipit peroksidasyonu, süperoksit dismutaz (SOD) aktivitesi, katalaz (CAT) aktivitesi, glutatyon peroksidaz (GPX) aktivitesi ölçülmüştür. Gruplara uygulanan ajanların sitotoksik dozları WST-1 testi ile bulunmuştur. Anneksin V ile apoptotik olmayan, erken apoptotik, geç apoptotik, toplam apoptotik ve ölü hücreler hem yüzde olarak hem de hücre/ml olarak hesaplanmıştır. Oksidatif stresle oluşan ve reaktif oksijen metabolitlerinin verdiği zararı belirlemek amacıyla lipit peroksidasyonu, SOD aktivitesi, CAT aktivitesi, GPX aktivitesi ölçülmüştür. İnsan yağ doku kaynaklı mezenkimal kök hücrelerde Sirolimus IC50 dozu (48. saat) için 15,85 μM olarak belirlenmiştir. UDKA için ise çalışılan dozlarda sitotoksik etki gözlemlenmemiştir. Bu yüzden en yüksek sabit doz olan 100 μM ile çalışmaya devam edilmiştir. Muse® hücre cihazı ile yapılan ölçümler ve yapılan hesaplamalar doğrultusunda kontrol grubunda ölü hücre %1.03, canlı hücre %98.97 olarak hesaplanmıştır. UDKA grubunda ölü hücreler %2.59, canlı hücreler %97.41 olarak hesaplanmıştır. Sirolimus grubunda erken apoptoz %7.69, geç apoptoz %4.31, ölü hücre %8.10, canlı hücre %79.90 olarak hesaplanmıştır. Sirolimus + UDKA kombine grupta erken apoptoz oranı %3.90, geç apoptoz oranı %1.34, ölü hücre %7.81, canlı hücre %86.95 olarak ölçülmüştür. Hücre siklusu analizinde kontrol ve UDKA grubunda G2/ M fazlarının yüzdeleri sırasıyla % 19.3 ve 18.9’ dur. Sirolimus uygulama grubunda G2/ M fazı yüzdesi 15.9 ’a düşmüştür. Biyokimyasal bulgular ise MDA düzeylerinde UDKA-kombine grubu; GSH-Px düzeyleri açısından Kontrol- UDKA dışındaki tüm ikili gruplar; CAT düzeyleri açısından kontrol- UDKA; kontrol- kombine; UDKA- Sirolimus ve UDKA- kombine grupları; SOD düzeyleri açısından tüm gruplar arası ikili farkların tümü istatistiksel olarak anlamlı olarak saptanmıştır (p, Mesenchymal stem cell therapies are important therapeutic modalities that have been applied experimentally and clinically in many fields of medicine today. Transplantation of mesenchymal stem cells into patients for supportive purposes besides immunosuppressive drugs is becoming common after organ transplantation in clinical cases. The aim of this study is to investigate the cytotoxic effects of Sirolimus, which is used for immunosuppressive purposes in organ transplants, on mesenchymal stem cells derivating from human adipose tissue and to investigate the effects of ursedeoxycholic acid (UDCA), which is known to have antioxidant properties, on this cytotoxicity. The human adipose tissue-derived mesenchymal stem cell line is commercially available. Human mesenchymal stem cells produced in special media which were incubated at 37 ° C until sufficient numbers were reached. The cytotoxic effects of the active substances were assessed by time and dose-dependent WST-1 test. Light microscopy and cell counting device were used for vitality and proliferation analyzes. Muse was used for evaluation of apoptosis and cell cycle in cell analyzer. Lipid peroxidation, superoxide dismutase (SOD) activity, catalase (CAT) activity, glutathione peroxidase (GPX) activity were measured by biochemical methods. Cytotoxic doses of the agents administered to the groups were found by the WST test. Non-apoptotic, early apoptotic, late apoptotic, total apoptotic and dead cells were calculated as both percent and cell/ml by Annexin V. Lipid peroxidation, SOD activity, CAT activity, GPX activity were measured to determine the damage caused by oxidative stress and reactive oxygen metabolites. In human adipose tissue-derived mesenchymal stem cells, Sirolimus IC50 was determined as 15.85 μM for dozen (48th hour). For UDCA, no cytotoxic effect was observed at the doses studied. Therefore, the study continued with the highest fixed dose of 100 μM. In the control group, according to the measurements made with Muse® cell analyzer the calculations made, the dead cell was calculated as 1.03% and the living cell as 98.97%. In the UDCA group, dead cells were calculated as 2.59% and living cells as 97.41%. In the Sirolimus group, early apoptosis was calculated as 7.69%, late apoptosis as 4.31%, dead cell as 8.10% and live cell as 79.90%. In the combined group of Sirolimus+ UDCA early apoptosis rate was 3.90%, late apoptosis rate was 1.34%, dead cell was 7.81% and viable cell was 86.95%. In the cell cycle analysis, the percentages of the G2/ M phases in the control and UDCA groups were 19.3% and 18.9%, respectively. The percentage of G2/ M phase in the Sirolimus treatment group was reduced to 15.9. Biochemical findings were; MDA- combined group; All binary groups except for Control- UDCA in terms of GSH-Px levels; Control in terms of CAT levels- UDCA; control-combined; UDCA- Sirolimus and UDCA- combined groups; SOD levels were statistically significant (p
- Published
- 2019
21. Propyl Gallate Inhibits Adipogenesis by Stimulating Extracellular Signal-Related Kinases in Human Adipose Tissue-Derived Mesenchymal Stem Cells
- Author
-
Ung-Kyu Choi, Seyoung Lim, Nan-Hee Lee, Jung-Min Kim, Jeung-Eun Lee, Seon-Jeong Choi, Hyun-Jun Jang, and Pann-Ghill Suh
- Subjects
MAPK/ERK pathway ,Adult ,Cellular differentiation ,Adipose tissue ,Biology ,chemistry.chemical_compound ,Adipocyte ,Lipid droplet ,Extracellular ,propyl gallate ,Humans ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Propyl gallate ,Cells, Cultured ,Adipogenesis ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,General Medicine ,Articles ,Cell biology ,chemistry ,Biochemistry ,Adipose Tissue ,Female ,human adipose tissue-derived mesenchymal stem cells - Abstract
Propyl gallate (PG) used as an additive in various foods has antioxidant and anti-inflammatory effects. Although the functional roles of PG in various cell types are well characterized, it is unknown whether PG has effect on stem cell differentiation. In this study, we demonstrated that PG could inhibit adipogenic differentiation in human adipose tissue-derived mesenchymal stem cells (hAMSCs) by decreasing the accumulation of intracellular lipid droplets. In addition, PG significantly reduced the expression of adipocyte-specific markers including peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAAT enhancer binding protein-α (C/EBP-α), lipoprotein lipase (LPL), and adipocyte fatty acid-binding protein 2 (aP2). PG inhibited adipogenesis in hAMSCs through extracellular regulated kinase (ERK) pathway. Decreased adipogenesis following PG treatment was recovered in response to ERK blocking. Taken together, these results suggest a novel effect of PG on adipocyte differentiation in hAMSCs, supporting a negative role of ERK1/2 pathway in adipogenic differentiation.
- Published
- 2015
22. Reversibility of hAT-MSCs phenotypic and metabolic changes after exposure to and withdrawal from HCC-conditioned medium through regulation of the ROS/MAPK/HIF-1α signaling pathway.
- Author
-
Wang, Chenyang, Hu, Jie, Chen, Zheng, Wang, Yifan, Lu, Sinan, Zhang, Yuan, Li, Yufeng, Xiang, Yucheng, Ji, Yutian, Zeng, Cheng, Ding, Yuan, and Wang, Weilin
- Subjects
CYCLIN-dependent kinases ,GLUCOSE metabolism ,MESENCHYMAL stem cells ,CELL morphology ,CELL cycle - Abstract
Background: Mesenchymal stem cells (MSCs) play an important role in tumor progression; concomitantly, MSCs also undergo profound changes in the tumor microenvironment (TME). These changes can directly impact the application and efficacy of MSC-based anti-tumor therapy. However, few studies have focused on the regulation of MSC fate in TME, which will limit the progress of MSC-based anti-tumor therapy. Herein, we investigated the effects of conditioned medium from human hepatocellular carcinoma cells (HCC-CM) on the phenotype and glucose metabolism of human adipose tissue-derived MSCs (hAT-MSCs). Methods: The passage 2 (P2) to passage 3 (P3) hAT-MSCs were exposed to conditioned medium from Hep3B, Huh7 and HCCLM3 cells for 4–8 weeks in vitro. Then, immunofluorescent, CCK-8 assay, EdU assay, Transwell assay, and flow cytometry were used to assess the alterations in cell phenotype in terms of cell morphology, secretory profiles, proliferation, migration, invasion, cell cycle, and apoptosis. In addition, glucose metabolism was evaluated by related kits. Next, the treated hAT-MSCs were subjected to withdrawal from HCC-CM for 2–4 weeks, and alterations in phenotype and glucose metabolism were reevaluated. Finally, the molecular mechanism was clarified by Western blotting. Results: The results revealed that after exposure to HCC-CM, hAT-MSCs developed a stellate-shaped morphology. In association with cytoskeleton remodeling, hAT-MSCs showed enhanced capacities for migration and invasion, while cell proliferation was inhibited by regulating the cell cycle by downregulating cyclins and cyclin-dependent kinases and activating the mitochondrial apoptosis pathway. In terms of glucose metabolism, our results showed mitochondrial dysfunction and elevated glycolysis of hAT-MSCs. However, interestingly, when the treated hAT-MSCs were subjected to withdrawal from HCC-CM, the alterations in phenotype and glucose metabolism could be reversed, but secretory phenotype and tumor-promoting properties appear to be permanent. Further studies showed that these changes in hAT-MSCs may be regulated by the ROS/MAPK/HIF-1α signaling pathway. Conclusion: Taken together, the effects of long-term HCC-CM treatment on phenotype and glucose metabolism in hAT-MSCs are modest and largely reversible after withdrawal, but HCC-CM endow hAT-MSCs with permanent secretory phenotype and tumor-promoting properties. This is the first report on the reversal of phenotype and glucose metabolism in tumor-associated MSCs (TA-MSCs), it is anticipated that new insights into TA-MSCs will lead to the development of novel strategies for MSC-based anti-tumor therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
23. PPARγ silencing enhances osteogenic differentiation of human adipose-derived mesenchymal stem cells
- Author
-
Gwo Jaw Wang, Hui Ting Chen, Mei-Ling Ho, Mon Juan Lee, Chung-Hwan Chen, Je Ken Chang, Lin Kang, Sung Cheng Huang, Shu-Chun Chuang, and Yin Chih Fu
- Subjects
Small interfering RNA ,medicine.medical_specialty ,Cellular differentiation ,Short Communications ,Peroxisome proliferator-activated receptor ,Biology ,Bone morphogenetic protein 2 ,peroxisome proliferator-activated receptor gamma ,osteogenesis ,Internal medicine ,medicine ,Humans ,Gene Silencing ,RNA, Small Interfering ,Cell Shape ,chemistry.chemical_classification ,Adipogenesis ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Osteoblast ,Cell Biology ,small interfering RNA ,Cell biology ,PPAR gamma ,RUNX2 ,medicine.anatomical_structure ,Endocrinology ,Adipose Tissue ,chemistry ,Molecular Medicine ,Human adipose tissue-derived mesenchymal stem cells - Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is the master regulator of adipogenesis, and has been indicated as a potential therapeutic target to promote osteoblast differentiation. However, recent studies suggest that suppression of PPARγ inhibits adipogenesis, but does not promote osteogenic differentiation in human bone marrow-derived mesenchymal stem cells (hBMSCs). It was reasoned that the osteogenic effect of PPARγ suppression may be masked by the strong osteogenesis-inducing condition commonly used, resulting in a high degree of matrix mineralization in both control and experimental groups. This study investigates the role of PPARγ in the lineage commitment of human adipose-derived mesenchymal stem cells (hADSCs) by interfering with the function of PPARγ mRNA through small interfering RNAs (siRNAs) specific for PPARγ2. By applying an osteogenic induction condition less potent than that used conventionally, we found that PPARγ silencing led to retardation of adipogenesis and stimulated a higher level of matrix mineralization. The mRNA level of PPARγ decreased to 47% of control 2 days after treatment with 50 nmol/l PPARγ2 siRNA, while its protein expression was 60% of mock control. In the meantime, osteogenic marker genes, including bone morphogenic protein 2 (BMP2), runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and osteocalcin (OC), were up-regulated under PPARγ silencing. Our results suggest that transient suppression of PPARγ promotes the onset of osteogenesis, and may be considered a new strategy to stimulate bone formation in bone tissue engineering using hADSCs.
- Published
- 2013
- Full Text
- View/download PDF
24. Alginate hydrogel enriched with enamel matrix derivative to target osteogenic cell differentiation in TiO2 scaffolds
- Author
-
Pullisaar, Helen, Verket, Anders, Szoke, Krisztina, Tiainen, Hanna, Haugen, Håvard J, Brinchmann, Jan E, Reseland, Janne E, and Østrup, Esben
- Subjects
primary human osteoblasts ,alginate hydrogel ,TiO2 scaffold ,human adipose tissue–derived mesenchymal stem cells ,Original Article ,Enamel matrix derivative - Abstract
The purpose of bone tissue engineering is to employ scaffolds, cells, and growth factors to facilitate healing of bone defects. The aim of this study was to assess the viability and osteogenic differentiation of primary human osteoblasts and adipose tissue-derived mesenchymal stem cells from various donors on titanium dioxide (TiO2) scaffolds coated with an alginate hydrogel enriched with enamel matrix derivative. Cells were harvested for quantitative reverse transcription polymerase chain reaction on days 14 and 21, and medium was collected on days 2, 14, and 21 for protein analyses. Neither coating with alginate hydrogel nor alginate hydrogel enriched with enamel matrix derivative induced a cytotoxic response. Enamel matrix derivative-enriched alginate hydrogel significantly increased the expression of osteoblast markers COL1A1, TNFRSF11B, and BGLAP and secretion of osteopontin in human osteoblasts, whereas osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells seemed unaffected by enamel matrix derivative. The alginate hydrogel coating procedure may have potential for local delivery of enamel matrix derivative and other stimulatory factors for use in bone tissue engineering.
- Published
- 2015
25. Establishment of Efficacy and Safety Assessment of Human Adipose Tissue-Derived Mesenchymal Stem Cells (hATMSCs) in a Nude Rat Femoral Segmental Defect Model
- Author
-
Sung Keun Kang, Jeong Chan Ra, Jong Min Kim, Hyung Jun Choi, Jeong Hwan Che, Byeong Cheol Kang, Seong Ryul Cho, Jae Il Lee, and Euna Kwon
- Subjects
Calcium Phosphates ,Male ,Pathology ,medicine.medical_specialty ,Bone Regeneration ,Cell Therapy & Organ Transplantation ,Efficacy ,Transplantation, Heterologous ,Adipose tissue ,Biocompatible Materials ,Human Adipose Tissue-Derived Mesenchymal Stem Cells ,HA/TCP Scaffold ,Bone healing ,Mesenchymal Stem Cell Transplantation ,Bone Defect Model ,Rats, Nude ,Tissue engineering ,Animals ,Humans ,Medicine ,Femur ,Bone regeneration ,Tissue Engineering ,business.industry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Histology ,General Medicine ,Rats ,Disease Models, Animal ,Durapatite ,Adipose Tissue ,Nude Rats ,Toxicity ,Original Article ,Diaphyses ,Bone Diseases ,Safety ,Tomography, X-Ray Computed ,business - Abstract
Human adipose tissue-derived mesenchymal stem cell (hATMSC) have emerged as a potentially powerful tool for bone repair, but an appropriate evaluation system has not been established. The purpose of this study was to establish a preclinical assessment system to evaluate the efficacy and safety of cell therapies in a nude rat bone defect model. Segmental defects (5 mm) were created in the femoral diaphyses and transplanted with cell media (control), hydroxyapatite/tricalcium phosphate scaffolds (HA/TCP, Group I), hATMSCs (Group II), or three cell-loading density of hATMSC-loaded HA/TCP (Group III-V). Healing response was evaluated by serial radiography, micro-computed tomography and histology at 16 weeks. To address safety-concerns, we conducted a GLP-compliant toxicity study. Scanning electron microscopy studies showed that hATMSCs filled the pores/surfaces of scaffolds in a cell-loading density-dependent manner. We detected significant increases in bone formation in the hATMSC-loaded HA/TCP groups compared with other groups. The amount of new bone formation increased with increases in loaded cell number. In a toxicity study, no significant hATMSC-related changes were found in body weights, clinical signs, hematological/biochemical values, organ weights, or histopathological findings. In conclusion, hATMSCs loaded on HA/TCP enhance the repair of bone defects and was found to be safe under our preclinical efficacy/safety hybrid assessment system.
- Published
- 2011
- Full Text
- View/download PDF
26. Propyl gallate inhibits adipogenesis by stimulating extracellular signal-related kinases in human adipose tissue-derived mesenchymal stem cells.
- Author
-
Lee JE, Kim JM, Jang HJ, Lim SY, Choi SJ, Lee NH, Suh PG, and Choi UK
- Subjects
- Adipose Tissue drug effects, Adipose Tissue enzymology, Adult, Cell Differentiation drug effects, Cells, Cultured, Female, Humans, Mesenchymal Stem Cells cytology, Adipogenesis drug effects, Adipose Tissue cytology, Extracellular Signal-Regulated MAP Kinases metabolism, Mesenchymal Stem Cells drug effects, Mesenchymal Stem Cells enzymology, Propyl Gallate pharmacology
- Abstract
Propyl gallate (PG) used as an additive in various foods has antioxidant and anti-inflammatory effects. Although the functional roles of PG in various cell types are well characterized, it is unknown whether PG has effect on stem cell differentiation. In this study, we demonstrated that PG could inhibit adipogenic differentiation in human adipose tissue-derived mesenchymal stem cells (hAMSCs) by decreasing the accumulation of intracellular lipid droplets. In addition, PG significantly reduced the expression of adipocyte-specific markers including peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAAT enhancer binding protein-α (C/EBP-α), lipoprotein lipase (LPL), and adipocyte fatty acid-binding protein 2 (aP2). PG inhibited adipogenesis in hAMSCs through extracellular regulated kinase (ERK) pathway. Decreased adipogenesis following PG treatment was recovered in response to ERK blocking. Taken together, these results suggest a novel effect of PG on adipocyte differentiation in hAMSCs, supporting a negative role of ERK1/2 pathway in adipogenic differentiation.
- Published
- 2015
- Full Text
- View/download PDF
27. Alginate hydrogel enriched with enamel matrix derivative to target osteogenic cell differentiation in TiO2 scaffolds.
- Author
-
Pullisaar H, Verket A, Szoke K, Tiainen H, Haugen HJ, Brinchmann JE, Reseland JE, and Østrup E
- Abstract
The purpose of bone tissue engineering is to employ scaffolds, cells, and growth factors to facilitate healing of bone defects. The aim of this study was to assess the viability and osteogenic differentiation of primary human osteoblasts and adipose tissue-derived mesenchymal stem cells from various donors on titanium dioxide (TiO2) scaffolds coated with an alginate hydrogel enriched with enamel matrix derivative. Cells were harvested for quantitative reverse transcription polymerase chain reaction on days 14 and 21, and medium was collected on days 2, 14, and 21 for protein analyses. Neither coating with alginate hydrogel nor alginate hydrogel enriched with enamel matrix derivative induced a cytotoxic response. Enamel matrix derivative-enriched alginate hydrogel significantly increased the expression of osteoblast markers COL1A1, TNFRSF11B, and BGLAP and secretion of osteopontin in human osteoblasts, whereas osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells seemed unaffected by enamel matrix derivative. The alginate hydrogel coating procedure may have potential for local delivery of enamel matrix derivative and other stimulatory factors for use in bone tissue engineering.
- Published
- 2015
- Full Text
- View/download PDF
28. Stem cells: insights into the secretome.
- Author
-
Makridakis M, Roubelakis MG, and Vlahou A
- Subjects
- Animals, Humans, Models, Molecular, Proteome analysis, Secretory Pathway, Proteome metabolism, Proteomics methods, Stem Cells cytology, Stem Cells metabolism
- Abstract
Stem cells have been considered as possible therapeutic vehicles for different health related problems such as cardiovascular and neurodegenerative diseases and cancer. Secreted molecules are key mediators in cell-cell interactions and influence the cross talk with the surrounding tissues. There is strong evidence supporting that crucial cellular functions such as proliferation, differentiation, communication and migration are strictly regulated from the cell secretome. The investigation of stem cell secretome is accumulating continuously increasing interest given the potential use of these cells in regenerative medicine. The scope of the review is to report the main findings from the investigation of stem cell secretome by the use of contemporary proteomics methods and discuss the current status of research in the field. This article is part of a Special Issue entitled: An Updated Secretome., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.