Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Med Assoc Thai ; 98 Suppl 3: S29-33, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26387385

RESUMO

BACKGROUND: Urinary leukotriene E4 (uLTE4) concentration represents body leukotriene synthesis. The level increases especially in respiratory allergic diseases. Researches of leukotriene production role in children with respiratory allergic diseases required the normal levels in non-allergic children as references. OBJECTIVE: The study was to assess the reference values of uLTE4 in non-allergic Thai children measured by competitive enzyme-linked immunosorbent assay (ELISA) technique. MATERIAL AND METHOD: Children who were admitted for elective surgery, aged 6 months to 5 years, were included in the study. Subjects who had acute illness, respiratory allergy (allergic rhinitis and asthma), and chronic diseases were excluded. Morning urine (5 ml) was collectedfor uLTE4 analysis by competitive ELISA technique. RESULTS: There were 36 urinary samples from 36 subjects aged 6-60 months with 26 boys and 10 girls. The mean of uLTE4 concentration was 619.73 ± 701.32 pg/ml and the mean of uLTE4 concentration adjustedfor urinary creatinine concentrations was 1,328 ± 788.54 pg/mg creatinine. The mean of uLTE4 concentration in boys and girls were 1,349 ± 817.10 pg/mg creatinine and 1,275 ± 747.79 pg/mg creatinine respectively. The mean of uLTE4 concentration adjusted for urinary creatinine concentrations were 1,363 ± 886.65, 1,384 ± 771.81 and 1,223 ± 773.16 pg/mg creatinine for children with the age of 6-18, 19-36 and 37-60 months respectively. CONCLUSION: The concentrations of uLTE4 and uLTE4 per creatinine were presented by age group in Thai very young children with non-respiratory disease. The concentrations were elevated in younger age group.


Assuntos
Leucotrieno E4/urina , Fatores Etários , Povo Asiático , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Lactente , Masculino , Valores de Referência
2.
J Med Assoc Thai ; 98 Suppl 3: S34-40, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26387386

RESUMO

Mesenchymal stem cells (MSCs) are multipotent stem cells which are able to differentiate into various lineages including osteoblasts, adipocytes and chondrocytes. They can be isolated from several tissues including bone marrow, adipose tissue, placenta and umbilical cord. Although MSCs could be diferentiated into osteoblasts under appropriate culture condition, their osteogenic differentiation capacity is still not very efficient. Previous studies reported that TNF-α could promote osteogenic differentiation of bone marrow derived MSCs by triggering NF-κB signaling pathway. However, the effect of TNF-α on the osteogenic differentiation ability ofumbilical cord derived MSCs has not been investigated. This study aimed to examine the effect of TNF-α on osteogenic differentiation of umbilical cord derived MSCs (UC-MSCs). The results demonstrated that TNF-α has osteopromotive effect for umbilical cord derived MSCs as evidenced by more matrix mineralization and alkaline phosphatase staining. Interestingly, UC-MSCs cultured in osteogenic differentiation medium supplemented with TNF-α had significantly increase expression of Osteocalcin, the marker of mature osteoblasts, when it was compared to UC-MSCs cultured in osteogenic differentiation medium without TNF-α (p < 0.05). On the contrary, the UC- MSCs cultured in osteogenic differentiation medium supplemented with TNF-α had significantly lower levels of Runx2 and Osterix (the markers of immature osteoblasts) than UC-MSCs cultured with osteogenic differentiation medium without TNF-α. The present study suggested that TNF-α promotes osteogenic differentiation of UC-MSCs. The data add a possibilityfor the use of UC-MSCs as an alternative source for cell replacement therapy in bone defect.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Cordão Umbilical/citologia , Tecido Adiposo/citologia , Células Cultivadas , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , NF-kappa B/metabolismo , Osteoblastos/citologia , Osteocalcina/metabolismo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
3.
Biochem Biophys Res Commun ; 447(4): 580-5, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24747566

RESUMO

Bortezomib (BZB) is a chemotherapeutic agent approved for treating multiple myeloma (MM) patients. In addition, there are several reports showing that bortezomib can induce murine mesenchymal stem cells (MSCs) to undergo osteogenic differentiation and increase bone formation in vivo. MSCs are the multipotent stem cells that have capacity to differentiate into several mesodermal derivatives including osteoblasts. Nowadays, MSCs mostly bone marrow derived have been considered as a valuable source of cell for tissue replacement therapy. In this study, the effect of bortezomib on the osteogenic differentiation of human MSCs derived from both bone marrow (BM-MSCs) and postnatal sources such as placenta (PL-MSCs) were investigated. The degree of osteogenic differentiation of BM-MSCs and PL-MSCs after bortezomib treatment was assessed by alkaline phosphatase (ALP) activity, matrix mineralization by Alizarin Red S staining and the expression profiles of osteogenic differentiation marker genes, Osterix, RUNX2 and BSP. The results showed that 1 nM and 2 nM BZB can induce osteogenic differentiation of BM-MSCs and PL-MSCs as demonstrated by increased ALP activity, increased matrix mineralization and up-regulation of osteogenic differentiation marker genes, Osterix, RUNX2 and BSP as compared to controls. The enhancement of osteogenic differentiation of MSCs by bortezomib may lead to the potential therapeutic applications in human diseases especially patients with osteopenia.


Assuntos
Ácidos Borônicos/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Placenta/citologia , Pirazinas/farmacologia , Adipogenia/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Doenças Ósseas Metabólicas/tratamento farmacológico , Bortezomib , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Feminino , Humanos , Sialoproteína de Ligação à Integrina/genética , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/genética , Osteogênese/fisiologia , Osteoporose/tratamento farmacológico , Gravidez , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Regulação para Cima/efeitos dos fármacos
4.
Cancer Cell Int ; 14: 69, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25788858

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is the most prevalent hepatic cancer worldwide. Currently, a targeted therapy via monoclonal antibodies (mAbs) specific to tumor-associated antigen is undergoing continual development in HCC treatment. METHODS: In this regard, after establishing and consequently exploring Hep88 mAb's tumoricidal effect on hepatocellular carcinoma cell line (HepG2 cell line), the Hep88 mAb's specific antigens from both membrane and cytoplasmic fractions of HepG2 cell line were identified by 2-D gel electrophoresis and western blot analysis. After in-gel digestion and subsequent analysis by liquid chromatography-mass spectrometry (LC-MS), mortalin (HSPA9) and alpha-enolase were identified. The recombinant proteins specific to Hep88 mAb were cloned and expressed in E. coli BL21(DE3). Moreover, alteration of HepG2 and Chang liver cell line after being induced by Hep88 mAb for 1-3 days was investigated using a transmission electron microscope. RESULTS: The result demonstrated that Hep88 mAb can bind to the recombinant mortalin (HSPA9) and alpha-enolase. In addition, the gradual appearing of mitochondria vacuolization and endoplasmic reticulum dilatation were observed. Those characteristics might be explained by the paraptosis-like program cell death (PCD), which is induced by the binding of Hep88 mAb to mortalin (HSPA9). Mortalin depletion resulting from the formation of Hep88 mAb-mortalin (HSPA9) complex might initiate transcription-independence of p53-mediated apoptosis. Additionally, Hep88mAb-alpha-enolase complex might initiate HepG2 cells energy exhaustion by glycolysis pathway obstruction. CONCLUSION: These fascinating results imply that Hep88 mAb might be a promising tool for the development of an effective treatment of HCC in the next decade.

5.
Biosci Rep ; 44(5)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38687607

RESUMO

Glioblastoma multiforme (GBM) is one of the most common and aggressive brain tumors. GBM resists most chemotherapeutic agents, resulting in a high mortality rate in patients. Human mesenchymal stem cells (hMSCs), which are parts of the cancer stroma, have been shown to be involved in the development and progression of GBM. However, different sources of hMSCs might affect GBM cells differently. In the present study, we established hMSCs from placenta (PL-hMSC) and chorion (CH-hMSC) to study the effects of their released soluble factors on the proliferation, migration, invasion, gene expression, and survival of human GBM cells, U251. We found that the soluble factors derived from CH-hMSCs and PL-hMSCs suppressed the proliferation of U251 cells in a dose-dependent manner. In contrast, soluble factors derived from both hMSC sources increased U251 migration without affecting their invasive property. The soluble factors derived from these hMSCs decreased the expression levels of CyclinD1, E2Fs and MYC genes that promote GBM cell proliferation but increased the expression level of TWIST gene, which promotes EMT and GBM cell migration. The functional study suggests that both hMSCs might exert their effects, at least in part, by activating TGF-ß and suppressing Wnt/ß-catenin signaling in U251 cells. Our study provides a better understanding of the interaction between GBM cells and gestational tissue-derived hMSCs. This knowledge might be used to develop safer and more effective stem cell therapy that improves the survival and quality of life of patients with GBM by manipulating the interaction between hMSCs and GBM cells.


Assuntos
Movimento Celular , Glioblastoma , Células-Tronco Mesenquimais , Fator de Crescimento Transformador beta , Via de Sinalização Wnt , Feminino , Humanos , Gravidez , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Proliferação de Células , Córion/metabolismo , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Glioblastoma/metabolismo , Glioblastoma/genética , Células-Tronco Mesenquimais/metabolismo , Placenta/metabolismo , Placenta/citologia , Fator de Crescimento Transformador beta/metabolismo
6.
Biosci Rep ; 2024 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-39364538

RESUMO

Fisetin and quercetin, polyphenol flavonoids, have been shown to have a wide range of beneficial pharmacological effects including anti-inflammatory, anti-oxidative, and anti-cancer. Our previous work shows that fisetin also affects the specification of the adipogenic-osteogenic lineage of human mesenchymal stem cells (hMSCs) by modulating the Hippo-YAP signaling pathway. Although quercetin has a structure similar to that of fisetin, its effects on the functional properties of hMSCs have not yet been investigated. The objective of this study is to determine the effects of quercetin on the various properties of hMSCs, including proliferation, migration, and differentiation capacity toward adipogenic and osteogenic lineages. The results show that while fisetin increases hMSC adipogenic differentiation, quercetin inhibited adipogenic differentiation of hMSCs. The inhibition is mediated, at least in part, by the activation of hippo signaling and up-regulation of miR-27b, which inhibits the expression of genes involved in all critical steps of lipid droplet biogenesis, resulting in a decrease in the number of lipid droplets in hMSCs. It is possible that the lack of hydroxylation of the 5 position on the A ring of quercetin could be responsible for its different effect on the adipogenic-osteogenic lineage specification of hMSCs compared to fisetin. Molecular docking and molecular dynamics simulation suggested that fisetin and quercetin possibly bind to serine / threonine protein kinases 4 (STK4/MST1), which is an upstream kinase responsible for LATS phosphorylation. Taken together, our results demonstrate more insight into the mechanism underlying the role of flavonoid fisetin and quercetin in the regulation of adipogenesis.

7.
J Med Assoc Thai ; 96(9): 1208-17, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24163998

RESUMO

OBJECTIVE: To explore the expression of pluripotent genes in Whartons jelly derived MSCs (WJ-MSCs) and their neuronal differentiation potential. MATERIAL AND METHOD: Gelatinous connective tissues from umbilical cord Wharton's jelly were digested with trypsin and then cultured in Dulbecco's Modified Eagle's Medium. The expressions of typical MSC markers as well as pluripotent markers were examined by flow cytometry and reverse transcription PCR, respectively. MSCs at passage 3 and 5 were used for in vitro adipogenic, osteogenic and neuronal differentiation by incubation with specific induction media. RESULTS: WJ-MSCs could be easily expanded for more than 20 passages while maintaining their undifferentiated state and their marker expression profiles, being positive for typical MSC markers CD90, CD73, and CD105, and being negative for hematopoietic markers CD34 and CD45. Interestingly, the expression of several pluripotent marker genes including Oct4, Rex1, Sox2, and Nanog was detected in early passages of both cultured WJ-MSCs and BM-MSCs. WJ-MSCs were able to differentiate not only to mesodermal cells, such as adipocyte and osteoblast but also the neural-like cells as characterized by neuronal morphology and the expression of neuronal markers including MAP-2, GFAP, beta-tubulin III and Tau. CONCLUSION: The present study demonstrates that WJ-MSCs can be readily obtained and expanded in culture while maintaining their typical MSC characteristics. WJ-MSCs and BM-MSCs also expressed several genes associated with pluripotency and exhibited their plasticity by differentiation toward neuronal-cell lineage. Umbilical cord Wharton's jelly might have potential to become an alternative source of MSC for treating nervous system disorders.


Assuntos
Expressão Gênica , Células-Tronco Mesenquimais/citologia , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/genética , Cordão Umbilical/citologia , Diferenciação Celular , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neurônios/citologia , Fotomicrografia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Geleia de Wharton
8.
Turk J Haematol ; 30(2): 115-21, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24385773

RESUMO

OBJECTIVE: 5-Azacytidine is a hypomethylating agent that is used for the treatment of myelodysplastic syndrome. This histone modifier is widely employed and plays a nonspecific role in influencing the differentiation capability of stem cells. The ability of bone marrow mesenchymal stem cells to differentiate into cardiomyocyte- and myocyte-like cells after exposure to 3 different doses of 5-azacytidine has been evaluated and compared. The aim of the study was to optimize the effective dose of 5-azacytidine for promoting the cardiomyocyte and myocyte differentiation capabilities of human mesenchymal stem cells (MSCs). MATERIALS AND METHODS: Human bone marrow aspirations were collected from healthy donors. MSCs were used for the study of mesodermal differentiation. MSCs were cultured to promote osteoblast differentiation and adipocyte differentiation. The evaluation of osteogenic or adipogenic properties was then performed through immunocytochemical staining. BMMSCs were trypsinized into single-cell suspensions and then prepared for flow cytometric analysis. The MSCs were treated with 5, 10, or 15 µM 5-azacytidine for 24 h and then cultured for 3 weeks. Total RNA was extracted from untreated and 5-azacytidine-treated cells. Troponin T and GATA4 antibodies were used as cardiogenic markers, whereas myogenin and MyoD antibodies were used as myocyte markers. RESULTS: The morphology and growth rate of MSCs that were treated with any of the 3 doses of 5-azacytidine were similar to the morphology and growth rate of control MSCs. An immunofluorescence analysis examining the expression of the cardiac-specific markers GATA4 and troponin T and the skeletal muscle-specific markers MyoD and myogenin revealed that cells treated with 15 µM 5-azacytidine were strongly positive for these markers. Real-time RT-PCR results were examined; these amplifications indicated that there were higher expression levels of cardiac- and skeletal muscle-specific mRNAs in MSCs treated with 15 µm 5-azacytidine than in MSCs that had either been treated with lower doses of 5-azacytidine or left untreated. CONCLUSION: MSCs treated with 5-azacytidine demonstrated the capacity to differentiate into both cardiomyocytes and skeletal myocytes, and 15 µM 5-azacytidine could be the optimal dose of this drug. Other promoting factors should be examined to investigate the possibility of promoting the differentiation of MSCs into specific cell types. CONFLICT OF INTEREST: None declared.

9.
Sci Rep ; 13(1): 22974, 2023 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-38151503

RESUMO

Placenta-derived mesenchymal stem cells (PL-MSCs) have therapeutic potential in various clinical contexts due to their regenerative and immunomodulatory properties. However, with increasing age or extensive in vitro culture, their viability and function are gradually lost, thus restricting their therapeutic application. The primary cause of this deterioration is oxidative injury from free radicals. Therefore, enhancing cell viability and restoring cellular repair mechanisms of PL-MSCs in an oxidative stress environment are crucial in this context. Fucoxanthin, a carotenoid derived from brown seaweed, demonstrates antioxidant activity by increasing the production of antioxidant enzymes and lowering the levels of reactive oxygen species (ROS). This study aimed to determine whether fucoxanthin protects PL-MSCs from hydrogen peroxide (H2O2)-induced oxidative stress. After characterization, PL-MSCs were co-treated with fucoxanthin and H2O2 for 24 h (co-treatment) or pre-treated with fucoxanthin for 24 h followed by H2O2 for 24 h (pre-treatment). The effects of fucoxanthin on cell viability and proliferation were examined using an MTT assay. The expression of antioxidant enzymes, PI3K/Akt/Nrf-2 and intracellular ROS production were investigated in fucoxanthin-treated PL-MSCs compared to the untreated group. The gene expression and involvement of specific pathways in the cytoprotective effect of fucoxanthin were investigated by high-throughput NanoString nCounter analysis. The results demonstrated that co-treatment and pre-treatment with fucoxanthin restored the viability and proliferative capacity of PL-MSCs. Fucoxanthin treatment increased the expression of antioxidant enzymes in PL-MSCs cultured under oxidative stress conditions and decreased intracellular ROS accumulation. Markedly, fucoxanthin treatment could restore PI3K/Akt/Nrf-2 expression in H2O2-treated PL-MSCs. High-throughput analysis revealed up-regulation of genes involved in cell survival pathways, including cell cycle and proliferation, DNA damage repair pathways, and down-regulation of genes in apoptosis and autophagy pathways. This study demonstrated that fucoxanthin protects and rescues PL-MSCs from oxidative stress damage through the PI3K/Akt/Nrf-2 pathway. Our data provide the supporting evidence for the use of fucoxanthin as an antioxidant cytoprotective agent to improve the viability and proliferation capacity of PL-MSCs both in vitro and in vivo required to increase the effectiveness of MSC expansion for therapeutic applications.


Assuntos
Antioxidantes , Células-Tronco Mesenquimais , Humanos , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Peróxido de Hidrogênio/toxicidade , Peróxido de Hidrogênio/metabolismo , Estresse Oxidativo , Apoptose
10.
Cells ; 12(24)2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-38132108

RESUMO

Mesenchymal stromal cells (MSCs) have recently been shown to play an important role in the growth and progression of many solid tumors, including cholangiocarcinoma (CCA). The human placental amniotic membrane (hPAM) is one of the most favorable sources of MSCs due to its availability and non-invasive harvesting procedure. However, the role of human placental amniotic membrane mesenchymal stromal cells (hPAMSCs) in the growth and progression of human CCA has not yet been determined. This study investigates the effects of conditioned medium derived from hPAMSCs (PA-CM) on the properties of three human CCA cell lines and explores possible mechanisms of action. Varying concentrations of PA-CM were used to treat CCA cells to determine their effects on the proliferation and apoptosis of CCA cells. The results showed that PA-CM inhibited the proliferation and colony-forming capacity of KKU100, KKU213A, and KKU213B cells. PA-CM also promoted the apoptosis of these CCA cells by causing the loss of mitochondrial membrane potential. Western Blotting confirmed that PA-CM induced CCA cell apoptosis by increasing the levels of the Bax/Bcl-2 ratio, cleaved caspase 3, and cleaved PARP, possibly by inhibiting the IL-6/JAK2/STAT3 signaling pathway. Moreover, our in vivo study also confirmed the suppressive effect of hPAMSCs on CCA cells by showing that PA-CM reduced tumor volume in nude mice transplanted with human CCA cells. Taken together, our results demonstrate that PA-CM has potent tumor-suppressive effects on human CCA cells and could potentially be used in combination with chemotherapy to develop a more effective treatment for CCA patients.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Células-Tronco Mesenquimais , Gravidez , Animais , Camundongos , Humanos , Feminino , Interleucina-6/metabolismo , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo , Âmnio/metabolismo , Camundongos Nus , Proliferação de Células , Placenta/metabolismo , Colangiocarcinoma/patologia , Transdução de Sinais , Ductos Biliares Intra-Hepáticos/patologia , Neoplasias dos Ductos Biliares/patologia , Apoptose , Células-Tronco Mesenquimais/metabolismo , Janus Quinase 2/metabolismo
11.
Ann Hematol ; 91(3): 321-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21808991

RESUMO

The origin of endothelial progenitor cells (EPCs) in umbilical cord blood (UCB) is unknown. In this study, we explored the origin of UCB-derived EPCs by culturing CD14+ or CD14- subpopulation separately and co-culturing these two subpopulations either with or without transwells. We found no colony formation with CD14+ or CD14- subpopulation alone, but there were EPC colonies observed in direct co-cultures of both subpopulations. Transwell culture system was used to further study the effect of cytokines on EPC colony formation. We observed the presence of EPC colonies derived from CD14- subpopulation in the presence of CD14+ subpopulation in the upper compartment whereas there was no colony generated from CD14+ subpopulation with CD14- subpopulation in the upper compartment. Therefore, CD14- subpopulation is likely to be the origin of EPCs and EPC colony derivation requires cytokines released from CD14+ subpopulation. We further characterized the founding population of UCB-derived EPCs by separating CD14- subpopulation into CD14-/CD34+ and CD14-/CD34- subpopulations. There were colonies observed only in co-cultures of CD14+ with CD14-/CD34+ subpopulation but not with CD14-/CD34- subpopulation either with or without transwells. We screened 42 cytokines involving in angiogenesis using an ELISA array in the supernatant collected from CD14+ compared to CD14- subpopulations. We found consistently the presence of angiogenin1 in the supernatant of CD14+ subpopulation but not in that of CD14- subpopulation. The addition of angiogenin1 in culture of CD14- subpopulation yielded EPC colonies. We conclude that UCB-derived EPCs are confined to CD14-/CD34+ subpopulation and angiogenin1 released from CD14+ subpopulation may be an important factor promoting the EPC colony formation.


Assuntos
Antígenos CD34/imunologia , Células Endoteliais/fisiologia , Sangue Fetal/citologia , Receptores de Lipopolissacarídeos/imunologia , Ribonuclease Pancreático/metabolismo , Células-Tronco/fisiologia , Antígenos CD34/genética , Separação Celular , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/citologia , Humanos , Recém-Nascido , Leucócitos Mononucleares/citologia , Receptores de Lipopolissacarídeos/genética , Células-Tronco/citologia
12.
Ann Hematol ; 91(3): 311-20, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21822618

RESUMO

In type 2 diabetes, the impairment of vascular repair processes and angiogenesis are due to endothelial progenitor cell (EPC) dysfunction. In this study, we established a quantitative methodology to assess EPC function by using an in vitro 5-(6)-carboxyfluorescein diacetate succinimidyl ester-labeling vessel formation assay. The EPCs were cultured in three different glucose concentrations (100, 189.5, and 295.5 mg/dl of D: -glucose) representing normal control and diabetes with either good or poor glycemic control, respectively. We found that the in vitro vessel-forming capacity was impaired in EPCs cultured in high glucose concentrations compared to normal control (43.4 ± 0.8% and 34.7 ± 0.7% vs. 50.8 ± 2.1%). We further studied expression of various genes involved in vessel formation. There was a lower level of angiopoietin 1 gene expression in EPCs cultured in high glucose concentrations. The addition of recombinant angiopoietin 1 significantly increased the vessel-forming capacity of EPCs cultured in high glucose concentration (35.3 ± 2.0% to 48.8 ± 2.7%), whereas the addition of angiopoietin 2 (a competitive inhibitor of angiopoietin 1) impaired the vessel-forming capacity of EPCs cultured in normal glucose concentration (51.8 ± 1.3% to 41.3 ± 0.6%). We conclude that the in vitro vessel-forming capacity of EPCs cultured in high glucose concentration is impaired due to low levels of angiopoietin 1.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Glucose/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Angiopoietina-1/farmacologia , Angiopoietina-2/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/fisiopatologia , Células Endoteliais/citologia , Fluoresceínas/metabolismo , Corantes Fluorescentes/metabolismo , Perfilação da Expressão Gênica , Humanos , Neovascularização Fisiológica/fisiologia , Células-Tronco/citologia , Succinimidas/metabolismo
13.
Stem Cells Int ; 2022: 4020845, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36406002

RESUMO

Background: Breast cancer is the most frequently diagnosed malignancy among women, resulting from abnormal proliferation of mammary epithelial cells. The highly vascularized nature of breast tissue leads to a high incidence of breast cancer metastases, resulting in a poor survival rate. Previous studies suggest that human mesenchymal stem cells (hMSCs) play essential roles in the growth, metastasis, and drug responses of many cancers, including breast cancer. However, hMSCs from different sources may release different combinations of cytokines that affect breast cancer differently. Methods: In this study, we have isolated hMSCs from the placenta (PL-hMSCs) and the chorion (CH-hMSCs) and determined how these hMSCs affect the proliferation, migration, invasion, and gene expression of two human breast cancer cells, MCF-7 and MDA-MB-231, as well as the possible mechanisms underlying those effects. Results: The results showed that the soluble factors derived from PL-hMSCs and CH-hMSCs inhibited the proliferation of MCF-7 and MDA-MB-231 cells but increased the migration of MDA-MB-231 cells. The study of gene expression showed that PL-hMSCs and CH-hMSCs downregulated the expression levels of the protooncogene CyclinD1 while upregulating the expression levels of tumor suppressor genes, P16 and P21 in MCF-7 and MDA-MB-231 cells. Furthermore, hMSCs from both sources also increased the expression levels of MYC, SNAI1, and TWIST, which promote the epithelial-mesenchymal transition and migration of breast cancer cells in both cell lines. The functional study suggests that the suppressive effect of CH-hMSCs and PL-hMSCs on MCF-7 and MDA-MB231 cell proliferation was mediated, at least in part, through IFN-γ. Conclusions: Our study suggests that CH-hMSCs and PL-hMSCs inhibited breast cancer cell proliferation by negatively regulating CYCLIND1 expression and upregulating the expression of the P16 and P21 genes. In contrast, hMSCs from both sources enhanced breast cancer cell migration, possibly by increasing the expression of MYC, SNAI1, and TWIST genes in those cells.

14.
Sci Rep ; 12(1): 19509, 2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-36376498

RESUMO

Mesenchymal stem cells (MSCs) are a promising candidate for bone repair. However, the maintenance of MSCs injected into the bone injury site remains inefficient. A potential approach is to develop a bone-liked platform that incorporates MSCs into a biocompatible 3D scaffold to facilitate bone grafting into the desired location. Bone tissue engineering is a multistep process that requires optimizing several variables, including the source of cells, osteogenic stimulation factors, and scaffold properties. This study aims to evaluate the proliferation and osteogenic differentiation potentials of MSCs cultured on 2 types of 3D-printed hydroxyapatite, including a 3D-printed HA and biomimetic calcium phosphate-coated 3D-printed HA. MSCs from bone marrow (BM-MSCs) and umbilical cord (UC-MSCs) were cultured on the 3D-printed HA and coated 3D-printed HA. Scanning electron microscopy and immunofluorescence staining were used to examine the characteristics and the attachment of MSCs to the scaffolds. Additionally, the cell proliferation was monitored, and the ability of cells to differentiate into osteoblast was assessed using alkaline phosphatase (ALP) activity and osteogenic gene expression. The BM-MSCs and UC-MSCs attached to a plastic culture plate with a spindle-shaped morphology exhibited an immunophenotype consistent with the characteristics of MSCs. Both MSC types could attach and survive on the 3D-printed HA and coated 3D-printed HA scaffolds. The MSCs cultured on these scaffolds displayed sufficient osteoblastic differentiation capacity, as evidenced by increased ALP activity and the expression of osteogenic genes and proteins compared to the control. Interestingly, MSCs grown on coated 3D-printed HA exhibited a higher ALP activity and osteogenic gene expression than those cultured on the 3D-printed HA. The finding indicated that BM-MSCs and UC-MSCs cultured on the 3D-printed HA and coated 3D-printed HA scaffolds could proliferate and differentiate into osteoblasts. Thus, the HA scaffolds could provide a suitable and favorable environment for the 3D culture of MSCs in bone tissue engineering. Additionally, biomimetic coating with octacalcium phosphate may improve the biocompatibility of the bone regeneration scaffold.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Osteogênese/genética , Durapatita/metabolismo , Medula Óssea , Alicerces Teciduais , Células Cultivadas , Diferenciação Celular/fisiologia , Cordão Umbilical , Proliferação de Células , Impressão Tridimensional
15.
Sci Rep ; 12(1): 11341, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35790790

RESUMO

Cholangiocarcinoma (CCA) is an aggressive malignancy arising from the damaged epithelial cells of the biliary tract. Previous studies have reported that the multi-potent mesenchymal stem cells (MSCs) activate a series of tumor signaling pathways by releasing several cytokines to influence tumor cell development. However, the roles and mechanisms of human chorion-derived MSCs (CH-MSCs) in cholangiocarcinoma progression have not been fully addressed. This present study aims to examine the effects of conditioned media derived from CH-MSCs (CH-CM) on CCA cell lines and investigate the respective underlying mechanism of action. For this purpose, MSCs were isolated from chorion tissue, and three cholangiocarcinoma cell lines, namely KKU100, KKU213A, and KKU213B, were used. MTT assay, annexin V/PI analysis, and JC-1 staining were used to assess the effects of CH-CM on proliferation and apoptosis of CCA cells, respectively. Moreover, the effect of CH-CM on caspase-dependent apoptotic pathways was also evaluated. The western blotting assay was also used for measuring the expression of JAK2/STAT3 signaling pathway-associated proteins. The results showed that CH-CM suppressed proliferation and promoted apoptosis of CCA cell lines. CH-CM treatment-induced loss of mitochondrial membrane potential (∆Ψm) in CCA cell lines. The factors presented in the CH-CM also inhibited JAK2/STAT3 signaling, reduced the expression of BCL-2, and increased BAX expression in CCA cells. In conclusion, our study suggests that the CH-CM has a potent anti-cancer effect on cholangiocarcinoma cells and thus provides opportunities for use in alternative cell therapy or in combination with a conventional chemotherapeutic drug to increase the efficiency of CCA treatment.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Células-Tronco Mesenquimais , Apoptose , Ductos Biliares Intra-Hepáticos , Linhagem Celular , Córion , Humanos , Fatores Imunológicos , Janus Quinase 2 , Neutropenia , Fator de Transcrição STAT3 , Transdução de Sinais
16.
J Med Assoc Thai ; 94 Suppl 7: S109-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22619916

RESUMO

Hep88 mAbs, a novel monoclonal antibodies against hepatocellular carcinoma cell line from Thai patient, has been proved earlier for its tumoricidal effect on HepG2 cell line. In the present study, we investigated not only Hep88 mAb's targeted proteins from HepG2 cell line by western blot analysis but also its inhibitory activity on those cells by MTT assay. Moreover the ultrastructural alteration induced by Hep88 mAb of HepG2 cell line compare with Chang liver cell line was also examined. The results demonstrated that Hep88 mAb had cytotoxic effect on HepG2 cell line but not Chang liver cell line. Additionally, recognizing proteins against Hep88 mAb have been found on both cell lines. The ultrastructural alteration detected from transmission electron microscopy included the appearing of intracellular vacuolization as well as the dilatation of endoplasmic reticulum and mitochondria have been observed. These findings are suggested that the death of HepG2 cell line after treatment with Hep88 mAb might be involved by an apoptosis-like program cell death (PCD) pathway. From all of these remarks, it is possible that Hep88 mAb can injure HCC cells by binding with its membrane-bound antigen and activated downstream intracellular signals which is finally leading cell to be death via apoptosis-like PCD.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Carcinoma Hepatocelular/ultraestrutura , Fatores Imunológicos/farmacologia , Neoplasias Hepáticas/ultraestrutura , Animais , Anticorpos Monoclonais/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/imunologia , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Células Hep G2/efeitos dos fármacos , Células Hep G2/ultraestrutura , Humanos , Fatores Imunológicos/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/imunologia , Camundongos , Camundongos Endogâmicos BALB C
17.
Sci Rep ; 11(1): 7670, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33828198

RESUMO

Mesenchymal stem cells (MSCs) are important in regenerative medicine because of their potential for multi-differentiation. Bone marrow, chorion and placenta have all been suggested as potential sources for clinical application. However, the osteogenic differentiation potential of MSCs derived from chorion or placenta is not very efficient. Bone morphogenetic protein-2 (BMP-2) plays an important role in bone development. Its effect on osteogenic augmentation has been addressed in several studies. Recent studies have also shown a relationship between miRNAs and osteogenesis. We hypothesized that miRNAs targeted to Runt-related transcription factor 2 (Runx-2), a major transcription factor of osteogenesis, are responsible for regulating the differentiation of MSCs into osteoblasts. This study examines the effect of BMP-2 on the osteogenic differentiation of MSCs isolated from chorion and placenta in comparison to bone marrow-derived MSCs and investigates the role of miRNAs in the osteogenic differentiation of MSCs from these sources. MSCs were isolated from human bone marrow, chorion and placenta. The osteogenic differentiation potential after BMP-2 treatment was examined using ALP staining, ALP activity assay, and osteogenic gene expression. Candidate miRNAs were selected and their expression levels during osteoblastic differentiation were examined using real-time RT-PCR. The role of these miRNAs in osteogenesis was investigated by transfection with specific miRNA inhibitors. The level of osteogenic differentiation was monitored after anti-miRNA treatment. MSCs isolated from chorion and placenta exhibited self-renewal capacity and multi-lineage differentiation potential similar to MSCs isolated from bone marrow. BMP-2 treated MSCs showed higher ALP levels and osteogenic gene expression compared to untreated MSCs. All investigated miRNAs (miR-31, miR-106a and miR148) were consistently downregulated during the process of osteogenic differentiation. After treatment with miRNA inhibitors, ALP activity and osteogenic gene expression increased over the time of osteogenic differentiation. BMP-2 has a positive effect on osteogenic differentiation of chorion- and placenta-derived MSCs. The inhibition of specific miRNAs enhanced the osteogenic differentiation capacity of various MSCs in culture and this strategy might be used to promote bone regeneration. However, further in vivo experiments are required to assess the validity of this approach.


Assuntos
Diferenciação Celular , Córion/citologia , Células-Tronco Mesenquimais/fisiologia , Osteogênese , Placenta/citologia , Fosfatase Alcalina/metabolismo , Proteína Morfogenética Óssea 2 , Regeneração Óssea , Feminino , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Gravidez
18.
J Agric Food Chem ; 69(32): 9259-9269, 2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34357771

RESUMO

Obesity has become a major public health concern; so, a strategy to prevent or reduce obesity is a priority. The inhibition of lipid droplet accumulation and adipogenesis process provides a target for the treatment of obesity. Herein, the effect of andrographolide (AP) on lipid accumulation in adipocytes derived from human bone marrow mesenchymal stem cells (hBM-MSCs) was examined. AP at concentrations of 1, 2.5, 5, and 10 µM reduced lipid droplet accumulation in the adipocytes by suppressing the adipogenic differentiation of hBM-MSCs. Concurrently, the expressions of adipogenic marker genes and the level of adipokines secreted by adipocytes were suppressed. Gene screening analysis showed a negative regulation of genes involved in the adipogenesis process. In conclusion, we demonstrated for the first time an antilipid accumulation in adipocytes from hBM-MSCs by AP. The compound may potentially be a novel therapeutic agent for the treatment of obesity as well as obesity-related diseases.


Assuntos
Adipogenia , Células-Tronco Mesenquimais , Adipócitos , Diferenciação Celular , Células Cultivadas , Diterpenos , Humanos , Gotículas Lipídicas
19.
Stem Cell Res Ther ; 12(1): 241, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33853681

RESUMO

INTRODUCTION: The in vitro expansion and differentiation of mesenchymal stem cells derived from bone marrow (BM-hMSCs) are considered as potential therapeutic tools for clinical applications in bone tissue engineering and regenerative medicine. However, invasive sampling and reduction in number and proliferative capacity with age are the major limitations of BM-hMSCs. Recently, human placenta-derived MSCs (PL-hMSCs) obtained by a non-invasive procedure have attracted much interest. Attempts to increase the potential of PL-hMSCs would be an important paradigm in regenerative medicine. Herein, we examined the proliferative and osteogenic effect of andrographolide (AP) on PL-hMSCs. METHODS: Mesenchymal stem cells were isolated from full-term normal human placentas and were characterized before using. Cell cytotoxicity and proliferative effect of AP were examined by MTT and BrdU assay, respectively. The non-toxicity concentrations of AP were further assessed for osteogenic effect determined by alkaline phosphatase (ALP) expression and activity, alizarin red staining, and osteoblast-specific gene expressions. Screening of genes involved in osteogenic differentiation-related pathways modulated by AP was explored by a NanoString nCounter analysis. RESULTS: PL-hMSCs generated in this study met the MSC criteria set by the International Society of Cellular Therapy. The non-cytotoxic concentrations of AP on PL-hMSCs are up to 10 µM. The compound increased PL-hMSC proliferation concomitant with increases in Wnt/ß-catenin level and activity. It also enhanced osteogenic differentiation in association with osteoblast-specific mRNA expression. Further, AP promoted bone formation and increased bone structural protein level, osteocalcin, in osteoblastic cells. Gene screening analysis showed the upregulation of genes related to Wnt/ß-catenin, TGFß/BMP, SMAD, and FGF signaling pathways. CONCLUSION: We demonstrated, for the first time, the potential role of AP in promoting proliferation, osteogenic differentiation, and osteoblast bone formation of PL-hMSCs. This study suggests that AP may be an effective novel agent for the improvement of PL-hMSCs and stem cell-based therapy for bone regeneration.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Diferenciação Celular , Células Cultivadas , Diterpenos , Feminino , Humanos , Placenta , Gravidez , beta Catenina
20.
BMC Endocr Disord ; 10: 5, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20374643

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) play an important role in vascular repair and a decrease in the number of EPCs is observed in type 2 diabetes. However, there is no report on the change of EPCs after glycemic control. This study therefore aimed to investigate the EPC number and function in patients with good and poor glycemic control. METHODS: The number of EPCs was studied using flow cytometry by co-expression of CD34 and VEGFR2. The EPCs were cultured and characterized by the expression of UEA-I, CD34, VEGFR2, vWF and Dil-Ac-LDL engulfment, as well as the ability to form capillary-like structures. An in vitro study on the effect of hyperglycemia on the proliferation and viability of the cultured EPCs was also performed. RESULTS: The number of EPCs in type 2 diabetes was significantly decreased compared with healthy controls and there was an inverse correlation between the EPC numbers and plasma glucose, as well as HbA1C. The number and function of EPCs in patients with good glycemic control were recovered compared with those with poor glycemic control. When glucose was supplemented in the culture in vitro, there was a negative effect on the proliferation and viability of EPCs, in a dose-dependent manner, whereas the enhancement of apoptosis was observed. CONCLUSION: There was EPC dysfunction in type 2 diabetes which might be improved by strict glycemic control. However, the circulating EPC number and proliferative function in patients with good glycemic control did not reach the level in healthy controls.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA