Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biosci Rep ; 44(5)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38687607

RESUMEN

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.


Asunto(s)
Movimiento Celular , Glioblastoma , Células Madre Mesenquimatosas , Factor de Crecimiento Transformador beta , Vía de Señalización Wnt , Femenino , Humanos , Embarazo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular , Corion/metabolismo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Glioblastoma/metabolismo , Glioblastoma/genética , Células Madre Mesenquimatosas/metabolismo , Placenta/metabolismo , Placenta/citología , Factor de Crecimiento Transformador beta/metabolismo
2.
Cells ; 12(24)2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38132108

RESUMEN

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.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Células Madre Mesenquimatosas , Embarazo , Animales , Ratones , Humanos , Femenino , Interleucina-6/metabolismo , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/metabolismo , Amnios/metabolismo , Ratones Desnudos , Proliferación Celular , Placenta/metabolismo , Colangiocarcinoma/patología , Transducción de Señal , Conductos Biliares Intrahepáticos/patología , Neoplasias de los Conductos Biliares/patología , Apoptosis , Células Madre Mesenquimatosas/metabolismo , Janus Quinasa 2/metabolismo
3.
J Nat Prod ; 86(5): 1335-1344, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37137165

RESUMEN

While obesity is a well-known health threatening condition worldwide, effective pharmacological interventions for obesity suppression have been limited due to adverse effects. Therefore, it is important to explore alternative medical treatments for combating obesity. Inhibition of the adipogenesis process and lipid accumulation are critical targets for controlling and treating obesity. Gardenia jasminoides Ellis is a traditional herbal remedy for various ailments. A natural product from its fruit, genipin, has major pharmacological properties; it is anti-inflammatory and antidiabetic. We investigated the effects of a genipin analogue, G300, on adipogenic differentiation in human bone marrow mesenchymal stem cells (hBM-MSCs). G300 suppressed the expression of adipogenic marker genes and adipokines secreted by adipocytes at concentrations of 10 and 20 µM, which effectively reduced the adipogenic differentiation of hBM-MSCs and lipid accumulation in adipocytes. It also improved adipocyte function by lowering inflammatory cytokine secretion and increasing glucose uptake. For the first time, we show that G300 has the potential to be a novel therapeutic agent for the treatment of obesity and its related disorders.


Asunto(s)
Adipogénesis , Células Madre Mesenquimatosas , Humanos , Médula Ósea/metabolismo , Células Cultivadas , Diferenciación Celular , Obesidad , Lípidos , Células de la Médula Ósea
4.
Sci Rep ; 12(1): 19509, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36376498

RESUMEN

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.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Humanos , Osteogénesis/genética , Durapatita/metabolismo , Médula Ósea , Andamios del Tejido , Células Cultivadas , Diferenciación Celular/fisiología , Cordón Umbilical , Proliferación Celular , Impresión Tridimensional
5.
Stem Cells Int ; 2022: 4020845, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36406002

RESUMEN

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.

6.
Sci Rep ; 12(1): 11341, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35790790

RESUMEN

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.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Células Madre Mesenquimatosas , Apoptosis , Conductos Biliares Intrahepáticos , Línea Celular , Corion , Humanos , Factores Inmunológicos , Janus Quinasa 2 , Neutropenia , Factor de Transcripción STAT3 , Transducción de Señal
7.
J Agric Food Chem ; 69(32): 9259-9269, 2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34357771

RESUMEN

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.


Asunto(s)
Adipogénesis , Células Madre Mesenquimatosas , Adipocitos , Diferenciación Celular , Células Cultivadas , Diterpenos , Humanos , Gotas Lipídicas
8.
Stem Cell Res Ther ; 12(1): 241, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33853681

RESUMEN

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.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Diferenciación Celular , Células Cultivadas , Diterpenos , Femenino , Humanos , Placenta , Embarazo , beta Catenina
9.
Sci Rep ; 11(1): 7670, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33828198

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Corion/citología , Células Madre Mesenquimatosas/fisiología , Osteogénesis , Placenta/citología , Fosfatasa Alcalina/metabolismo , Proteína Morfogenética Ósea 2 , Regeneración Ósea , Femenino , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Embarazo
10.
Biomed Pharmacother ; 131: 110763, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33152926

RESUMEN

A decline of estrogen in menopause women is accompanied with increases in many pro-inflammatory cytokines and osteoporosis. Andrographolide (AP), from Andrographis paniculata, which has an anti-inflammatory activity, may have potential to alleviate osteoporosis during estrogen deficiency. Here we report the promoting effects of AP on the differentiation of mouse pre-osteoblastic (MC3T3-E1) cells by increasing the expression and activity of alkaline phosphatase (ALP), an osteoblastic gene-specific marker. AP also accelerated bone formation and increased bone structural gene production including collagen and osteocalcin. We demonstrate for the first time the promoting effect of AP on the differentiation of osteoblast involved with the OPG/RANKL signaling pathway. AP also protected bone loss in the estrogen-deficient (ovariectomized, OVX) rats after 12 weeks of treatment. It protected the loss of bone mineral density, bone microarchitecture, and improved bone turnover rate in OVX rats. This study provides an essential evidence for clinical applications and development of AP towards treating osteoporosis in post-menopausal women.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diterpenos/farmacología , Osteoblastos/efectos de los fármacos , Osteoporosis Posmenopáusica/prevención & control , Células 3T3 , Andrographis/química , Animales , Densidad Ósea/efectos de los fármacos , Modelos Animales de Enfermedad , Estrógenos/deficiencia , Femenino , Humanos , Ratones , Osteoblastos/citología , Osteogénesis/efectos de los fármacos , Osteoprotegerina/metabolismo , Ovariectomía , Ligando RANK/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Stem Cells Int ; 2019: 9748795, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31354842

RESUMEN

BACKGROUND: Cancer has been considered a serious global health problem and a leading cause of morbidity and mortality worldwide. Despite recent advances in cancer therapy, treatments of advance stage cancers are mostly ineffective resulting in poor survival of patients. Recent evidences suggest that multipotent human mesenchymal stem cells (hMSCs) play important roles in growth and metastasis of several cancers by enhancing their engraftment and inducing tumor neovascularization. However, the effect of hMSCs on cancer cells is still controversial because there are also evidences demonstrating that hMSCs inhibited growth and metastasis of some cancers. METHODS: In this study, we investigated the effects of bioactive molecules released from bone marrow and gestational tissue-derived hMSCs on the proliferation of various human cancer cells, including C3A, HT29, A549, Saos-2, and U251. We also characterized the hMSC-derived factors that inhibit cancer cell proliferation by protein fractionation and mass spectrometry analysis. RESULTS: We herein make a direct comparison and show that the effects of hMSCs on cancer cell proliferation and migration depend on both hMSC sources and cancer cell types and cancer-derived bioactive molecules did not affect the cancer suppressive capacity of hMSCs. Moreover, hMSCs use distinct combination of bioactive molecules to suppress the proliferation of human hepatoblastoma and colorectal cancer cells. Using protein fractionation and mass spectrometry analysis, we have identified several novel hMSC-derived factors that might be able to suppress cancer cell proliferation. CONCLUSION: We believe that the procedure developed in this study could be used to discover other therapeutically useful molecules released by various hMSC sources for a future in vivo study.

12.
Stem Cell Res Ther ; 10(1): 79, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30845980

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) are considered potential candidates that hold great promise in the treatment of immune-related diseases. For therapeutic applications, it is necessary to isolate and expand MSCs with procedures complying with good manufacturing practice (GMP). Recent studies reported the use of human serum (HS) instead of fetal bovine serum (FBS) for the expansion of bone marrow-derived MSCs. Nevertheless, there are only limited data on HS as an alternative to FBS for the isolation and expansion of umbilical (UC-MSCs) and placenta-derived MSCs (PL-MSCs). In this study, we evaluate the effect of HS compared to FBS on the proliferative and immunosuppressive capacities of these MSCs. METHODS: PL-MSCs and UC-MSCs were isolated and cultured in HS- or FBS-supplemented media. The MSC characteristics, including morphology, immunophenotype, and differentiation ability, were verified. The proliferative and immunosuppressive capacities were also examined. In addition, the proliferative-enhancing factors in both sera were explored using proteomic analysis. RESULTS: PL-MSCs and UC-MSCs proliferated faster in HS-supplemented medium than in equivalent levels of FBS-supplemented medium. Adipogenic and osteogenic differentiations occurred at nearly identical levels in HS- and FBS-supplemented media. Interestingly, MSCs cultured in HS-supplemented medium had a similar immunosuppressive effect as MSCs cultured in FBS-supplemented medium. Proteomic analysis revealed that Con-A binding glycoproteins with a molecular weight > 100 kDa in FBS could significantly enhance MSC proliferation. In contrast, the proliferative enhancing factors in HS were found in the Con-A non-binding fraction and WGA binding fraction with a molecular weight > 100 kDa. CONCLUSIONS: Taken together, our results suggest applications for the use of HS instead of FBS for the isolation and expansion of PL-MSCs and UC-MSCs for cell therapy in the future. Furthermore, this study identifies factors in HS that are responsible for its proliferative and immunosuppressive effects and might thus lead to the establishment of GMPs for the therapeutic use of MSCs.


Asunto(s)
Proliferación Celular , Inmunomodulación , Células Madre Mesenquimatosas/inmunología , Placenta/inmunología , Suero , Cordón Umbilical/inmunología , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Placenta/citología , Embarazo , Cordón Umbilical/citología
13.
JBMR Plus ; 2(4): 217-226, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30123862

RESUMEN

Production of sphingosine-1-phosphate (S1P) is linked to 17ß-estradiol (E2) activity in many estrogen-responsive cells; in bone development, the role of S1P is unclear. We studied effects of S1P on proliferation and differentiation of human osteoblasts (hOB). Ten nM E2, 1 µM S1P, or 1 µM of the S1P receptor 1 (S1PR1) agonist SEW2871 increased hOB proliferation at 24 hours. S1PR 1, 2, and 3 mRNAs are expressed by hOB but not S1PR4 or S1PR5. Expression of S1PR2 was increased at 7 and 14 days of differentiation, in correspondence with osteoblast-related mRNAs. Expression of S1PR1 was increased by E2 or S1P in proliferating hOB, whereas S1PR2 mRNA was unaffected in proliferating cells; S1PR3 was not affected by E2 or S1P. Inhibiting sphingosine kinase (SPHK) activity with sphingosine kinase inhibitor (Ski) greatly reduced the E2 proliferative effect. Both E2 and S1P increased SPHK mRNA at 24 hours in hOB. S1P promoted osteoblast proliferation via activating MAP kinase activity. Either E2 or S1P increased S1P synthesis in a fluorescent S1P assay. Interaction of E2 and S1P signaling was indicated by upregulation of E2 receptor mRNA after S1P treatment. E2 and S1P also promoted alkaline phosphatase expression. During osteoblast differentiation, S1P increased bone-specific mRNAs, similarly to the effects of E2. However, E2 and S1P showed differences in the activation of some osteoblast pathways. Pathway analysis by gene expression arrays was consistent with regulation of pathways of osteoblast differentiation; collagen and cell adhesion proteins centered on Rho/Rac small GTPase signaling and Map kinase or signal transducer and activator of transcription (Stat) intermediates. Transcriptional activation also included significant increases in superoxide dismutase 1 and 2 transcription by either S1P or E2. We demonstrate that the SPHK system is a co-mediator for osteoblast proliferation and differentiation, which is mainly, but not entirely, complementary to E2, whose effects are mediated by S1PR1 and S1PR2.

14.
Int J Mol Med ; 39(3): 654-662, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28204808

RESUMEN

Mesenchymal stromal cells (MSCs) are multipotent cells that can give rise to different cell types of the mesodermal lineages. They are powerful sources for cell therapy in regenerative medicine as they can be isolated from various tissues, and can be expanded and induced to differentiate into multiple lineages. Recently, the umbilical cord has been suggested as an alternative source of MSCs. Although MSCs derived from the umbilical cord can be induced to differentiate into osteoblasts with a phenotypic similarity to that of bone marrow-derived MSCs, the differentiation ability is not consistent. In addition, MSCs from the umbilical cord require a longer period of time to differentiate into osteoblasts. Previous studies have demonstrated the benefits of bone morphogenetic protein-2 (BMP-2) in bone tissue regeneration. In addition, several studies have supported the use of BMP-2 in periodontal regeneration, sinus lift bone-grafting and non-unions in oral surgery. Although the use of BMP-2 for bone tissue regeneration has been extensively investigated, the BMP-2-induced osteogenic differentiation of MSCs derived from the umbilical cord has not yet been fully examined. Therefore, in this study, we aimed to examine the effects of BMP-2 on the osteogenic differentiation of MSCs derived from umbilical cord compared to that of MSCs derived from bone marrow. The degree of osteogenic differentiation following BMP-2 treatment was determined by assessing alkaline phosphatase (ALP) activity, and the expression profiles of osteogenic differentiation marker genes, osterix (Osx), Runt-related transcription factor 2 (Runx2) and osteocalcin (Ocn). The results revealed that BMP-2 enhanced the osteogenic differentiation capacity of MSCs derived from both bone marrow and umbilical cord as demonstrated by increased ALP activity and the upregulation of osteogenic differentiation marker genes. The enhancement of the osteogenic differentiation capacity of MSCs by BMP-2 suggests that these MSCs may be used as alternative sources for bone engineering or cell therapy in regenerative medicine.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Adipogénesis/efectos de los fármacos , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Fenotipo
15.
Stem Cells Int ; 2017: 7257628, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29348760

RESUMEN

Mesenchymal stromal cells (MSCs) offering valuable anticipations for the treatment of degenerative diseases. They can be found in many tissues including amnion. MSCs from amnion (AM-MSCs) can differentiate into osteoblast similar to that of bone marrow-derived MSCs (BM-MSCs). However, the ability is not much efficient compared to BM-MSCs. This study aimed to examine the effects of BMP-2 and miRNAs on osteogenic differentiation of AM-MSCs compared to those of BM-MSCs. The osteogenic differentiation capacity after miRNA treatment was assessed by ALP expression, ALP activity, and osteogenic marker gene expression. The results showed that the osteogenic differentiation capacity increased after BMP-2 treatment both in AM-MSCs and BM-MSCs. MiR-31, miR-106a, and miR-148a were downregulated during the osteogenic differentiation. After transfection with anti-miRNAs, ALP activity and osteogenic genes were increased over the time of differentiation. The data lead to the potential for using AM-MSCs as an alternative source for bone regeneration. Moreover, the information of miRNA expression and function during osteogenic differentiation may be useful for the development of new therapeutics or enhanced an in vitro culture technique required for stem cell-based therapies in the bone regeneration.

16.
Stem Cells Int ; 2016: 9674614, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27057179

RESUMEN

Most type 2 diabetic patients are obese who have increased number of visceral adipocytes. Those visceral adipocytes release several factors that enhance insulin resistance making diabetic treatment ineffective. It is known that significant percentages of visceral adipocytes are derived from mesenchymal stem cells and high glucose enhances adipogenic differentiation of mouse bone marrow-derived MSCs (BM-MSCs). However, the effect of high glucose on adipogenic differentiation of human bone marrow and gestational tissue-derived MSCs is still poorly characterized. This study aims to investigate the effects of high glucose on proliferation as well as adipogenic and osteogenic differentiation of human MSCs derived from bone marrow and several gestational tissues including chorion, placenta, and umbilical cord. We found that high glucose reduced proliferation but enhanced adipogenic differentiation of all MSCs examined. The expression levels of some adipogenic genes were also upregulated when MSCs were cultured in high glucose. Although high glucose transiently downregulated the expression levels of some osteogenic genes examined, its effect on the osteogenic differentiation levels of the MSCs is not clearly demonstrated. The knowledge gained from this study will increase our understanding about the effect of high glucose on adipogenic differentiation of MSCs and might lead to an improvement in the diabetic treatment in the future.

17.
J Med Assoc Thai ; 99 Suppl 4: S75-83, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29917345

RESUMEN

Objective: Evidence exists indicating that mesenchymal stem cells (MSCs) are promising candidate for therapeutic applications. One major obstacle for their clinical use is the biosafety of fetal bovine serum (FBS), which is a crucial part of all media currently used for culture of MSCs. Although some recent studies recommended substituting FBS with human serum (HS) for the expansion of MSCs for clinical use, the characteristics and functional capacity of the expanded cells has only been partially explored. In addition, limited experience indicates that HS may replace FBS in some but not all culture systems. Currently, relatively little is known about using HS instead of FBS for isolation and expansion of placenta derived MSCs. Therefore, this study aimed to compare the exploit of HS and FBS as a supplement in terms of their impact on biological characteristics of MSCs. Material and Method: MSCs derived from placenta were cultured in Dulbecco's Modified Eagle's Medium supplemented with 10% fetal bovine serum or 10% human serum. The morphology, the expression of MSC markers, the differentiation ability and the proliferation characteristics were examined. Results: The results demonstrated that MSCs cultured in DMEM supplemented with 10% HS had similar characteristics to MSCs cultured in DMEM supplemented with 10% FBS. Interestingly, MSCs cultured in DMEM supplemented with 10% HS had greater expansion potential than that of MSCs cultured in DMEM supplemented with 10% FBS. Conclusion: The results obtained from this study imply some application in the use of HS instead of FBS for expansion of placenta derived MSCs. The HS-expanded MSCs might be useful and safe for use as a therapeutic tool in regenerative medicine.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Placenta/metabolismo , Suero/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Medio de Cultivo Libre de Suero , Femenino , Humanos , Embarazo
18.
J Med Assoc Thai ; 98 Suppl 3: S34-40, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26387386

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Cordón Umbilical/citología , Tejido Adiposo/citología , Células Cultivadas , Femenino , Humanos , Células Madre Mesenquimatosas/citología , FN-kappa B/metabolismo , Osteoblastos/citología , Osteocalcina/metabolismo , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
19.
PLoS One ; 8(11): e78739, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244350

RESUMEN

Phytoestrogens have been implicated in the prevention of bone loss in postmenopausal osteoporosis. Recently, an active phytoestrogen from Curcuma comosa Roxb, diarylheptanoid (DPHD), (3R)-1,7-diphenyl-(4E,6E)-4,6-heptadien-3-ol, was found to strongly promote human osteoblast function in vitro. In the present study, we demonstrated the protective effect of DPHD on ovariectomy-induced bone loss (OVX) in adult female Sprague-Dawley rats with 17ß-estradiol (E2, 10 µg/kg Bw) as a positive control. Treatment of OVX animals with DPHD at 25, 50, and 100 mg/kg Bw for 12 weeks markedly increased bone mineral density (BMD) of tibial metaphysis as measured by peripheral Quantitative Computed Tomography (pQCT). Histomorphometric analysis of bone structure indicated that DPHD treatment retarded the ovariectomy-induced deterioration of bone microstructure. Ovariectomy resulted in a marked decrease in trabecular bone volume, number and thickness and these changes were inhibited by DPHD treatment, similar to that seen with E2. Moreover, DPHD decreased markers of bone turnover, including osteocalcin and tartrate resistant acid phosphatase (TRAP) activity. These results suggest that DPHD has a bone sparing effect in ovariectomy-induced trabecular bone loss and prevents deterioration of bone microarchitecture by suppressing the rate of bone turnover. Therefore, DPHD appears to be a promising candidate for preserving bone mass and structure in the estrogen deficient women with a potential role in reducing postmenopausal osteoporosis.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Curcuma/química , Diarilheptanoides/farmacología , Osteoporosis Posmenopáusica/prevención & control , Ovariectomía , Fitoestrógenos/farmacología , Animales , Conservadores de la Densidad Ósea/química , Diarilheptanoides/química , Estradiol/farmacología , Femenino , Humanos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoporosis Posmenopáusica/metabolismo , Osteoporosis Posmenopáusica/patología , Fitoestrógenos/química , Ratas , Ratas Sprague-Dawley
20.
Phytomedicine ; 20(8-9): 676-82, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23557993

RESUMEN

Curcuma comosa Roxb. is ginger-family plant used to relieve menopausal symptoms. Previous work showed that C. comosa extracts protect mice from ovariectomy-induced osteopenia with minimal effects on reproductive organs, and identified the diarylheptanoid (3R)-1,7-diphenyl-(4E,6E)-4,6-heptadien-3-ol (DPHD) as the major active component of C. comosa rhizomes. At 1-10µM, DPHD increased differentiation in transformed mouse osteoblasts, but the effect of DPHD on normal bone cells was unknown. We examined the concentration dependency and mechanism of action of DPHD relative to 17ß-estradiol in nontransformed human osteoblasts (h-OB). The h-OB were 10-100 fold more sensitive to DPHD than transformed osteoblasts: DPHD increased h-OB proliferation at 10nM and, at 100nM, activated MAP kinase signaling within 30 min. In long-term differentiation assays, responses of h-OB to DPHD were significant at 10nM, and optimal response in most cases was at 100 nM. At 7-21 days, DPHD accelerated osteoblast differentiation, indicated by alkaline phosphatase activity and osteoblast-specific mRNA production. Effects of DPHD were eliminated by the estrogen receptor antagonist ICI182780. During differentiation, DPHD promoted early expression of osteoblast transcription factors, RUNX2 and osterix. Subsequently, DPHD accelerated production of bone structural genes, including COL1A1 and osteocalcin comparably to 17ß-estradiol. In h-OB, DPHD increased the osteoprotegerin to RANKL ratio and supported mineralization more efficiently than 10nM 17ß-estradiol. We conclude that DPHD promotes human osteoblast function in vitro effectively at nanomolar concentrations, making it a promising compound to protect bone in menopausal women.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Curcuma/química , Osteoblastos/efectos de los fármacos , Fitoestrógenos/farmacología , Extractos Vegetales/farmacología , Diarilheptanoides/química , Diarilheptanoides/metabolismo , Diarilheptanoides/farmacología , Estradiol/química , Estradiol/farmacología , Femenino , Heptanol/análogos & derivados , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Menopausia/efectos de los fármacos , Osteocalcina/metabolismo , Osteocalcina/farmacología , Osteoporosis/tratamiento farmacológico , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Fitoestrógenos/química , Fitoestrógenos/aislamiento & purificación , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , Rizoma/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...