scholarly journals Mesenchymal stem cells derived from breast cancer tissue promote the proliferation and migration of the MCF-7 cell line in vitro

2013 ◽  
Vol 6 (6) ◽  
pp. 1577-1582 ◽  
Author(s):  
CHUNFU ZHANG ◽  
WEI ZHAI ◽  
YAN XIE ◽  
QIAOLIN CHEN ◽  
WEI ZHU ◽  
...  
Author(s):  
Ana Catarina Viana Valle ◽  
Lana Ribeiro Aguiar ◽  
Hilana dos Santos Sena Brunel ◽  
Patricia Furtado Malard ◽  
Carla Lujan Pereira Villarroel ◽  
...  

BACKGROUND Adenocarcinomas can be of several types, and MCF-7 is an adenocarcinoma of human breast cell line useful as preclinical model to screen therapeutic agents such as ultra-diluted Viscum album, an European plant whose extract is commonly used in cancer therapy. AIMS MCF-7 and mesenchymal stem cells were used to evaluate the in vitro cytotoxicity of homoeopathic Viscum album 1x10-3 (VAD3). METHODS cells were cultured for 24 hours in controlled environment (37.5oC and 5% CO2) in 96-well plates. After this time, VAD3 was added to the culture medium in concentrations varying from 10 to 100 ?L/mL for MTT assay (evaluation of viability of cells). A control group was maintained with culture medium only. After 48 hours, the procedures of analysis of cells viability were performed. RESULTS MTT assay showed that the concentrations of 42 ?L/mL and 62 ?L/mL were able to reduce cell viability to 50% in MCF-7 and mesenchymal stem cells, respectively, which means that half of the cells cultured were dead after 48 hours in contact with VAD3. CONCLUSION Viscum album presented higher cytotoxic action on human breast cancer cell line culture than on mesenchymal stem cells. This medicine is extensively used against cancer, and the use of the homoeopathic form of it brings new possibilities as no or fewer adverse effects would be present.


2016 ◽  
Author(s):  
Anton J. Lucanus ◽  
Victoria King ◽  
George W. Yip

ABSTRACTBreast cancer pathogenesis is known to be propagated by the differential expression of a group of proteins called the Kinesin Superfamily (KIFs), which are instrumental in the intracellular transport of chromosomes along microtubules during mitosis. During mitosis, KIFs are strictly regulated through temporal synthesis so that they are only present when needed. However, their misregulation may contribute to uncontrolled cell growth due to premature sister chromatid separation, highlighting their involvement in tumorigenesis. One particular KIF, KIF21A, was recently found to promote the survival of human breast cancer cells in vitro. However, how KIF21A influences other cancerous phenotypes is currently unknown. This study therefore aimed to consolidate the in vitro role of KIF21A in breast cancer metastasis, while also analysing KIF21A expression in human breast cancer tissue to determine its prognostic value. This was achieved by silencing KIF21A in MCF-7 and MDA-MB-231 breast cancer cell lines via siRNA transfection. Migration, invasion, proliferation, and adhesion assays were then performed to measure the effects of KIF21A silencing on oncogenic behaviour. Immunohistochemistry was also conducted in 263 breast cancer tissue samples to compare KIF21A expression levels against various prognostic outcomes and clinicopathological parameters. KIF21A knockdown reduced cell migration (by 42.8% [MCF-7] and 69.7% [MDA-MB-231]) and invasion (by 72.5% [MCF-7] and 42.5% [MDA-MB-231]) in both cell lines, but had no effect on adhesion or proliferation, suggesting that KIF21A plays an important role in the early stages of breast cancer metastasis. Unexpectedly however, KIF21A was shown to negatively correlate with various pro-malignant clinicopathological parameters, including tumour size and histological grade, and high KIF21A expression predicted better breast cancer survival (hazard ratio = 0.45), suggesting that KIF21A is a tumour suppressor. The conflicting outcomes of in vitro and in vivo data may be due to the possible multi-functionality of KIF21A or study limitations, and means no definitive conclusions can be drawn about the role of KIF21A in breast cancer. This warrants further investigation, which may prove pivotal to the development of novel chemotherapeutic strategies to mediate KIF21A’s function and enhance prognostic outcomes.


2016 ◽  
Vol 308 (7) ◽  
pp. 511-520 ◽  
Author(s):  
Talita Stessuk ◽  
Maria Beatriz Puzzi ◽  
Elinton Adami Chaim ◽  
Paulo César Martins Alves ◽  
Erich Vinicius de Paula ◽  
...  

2018 ◽  
Vol 46 (1) ◽  
pp. 279-290 ◽  
Author(s):  
Dongye Yi ◽  
Wei Xiang ◽  
Qing Zhang ◽  
Yongcun Cen ◽  
Qing Su ◽  
...  

Background/Aims: Tumor vascular formation and maintenance are crucial events in glioblastoma development. Mesenchymal stem cells (MSCs) have been shown to differentiate into pericytes and contribute to neovascularization in the glioma microenvironment. Moreover, glioblastoma-derived mesenchymal stem cells (gb-MSCs), which consist of CD90-MSCs and CD90+MSCs, are a subpopulation of MSCs that are more active in glioma vascularization. However, the functions of gb-MSCs and the microRNA (miRNA) modifications in the glioblastoma microenvironment have not yet been fully elucidated. Here, we focus on the pericyte differentiation potential of gb-MSCs and miRNA modifications in gb-MSCs during new vascular formation and glioblastoma growth. Methods: In vitro, surface markers of gb-MSCs were detected by flow cytometry; the differentiation potential was evaluated by Oil Red O staining, Alizarin Red staining and Alcian blue staining; the proliferation and migration of gb-MSCs in different conditioned media were analyzed by the cck8 test and wound-healing assay, respectively; gb-MSC to pericyte transition was detected by immunofluorescence staining and western blot assay; angiogenetic capacity was analyzed by tube formation assay; and levels of cytokines in different supernatant were determined by ELISA. Additionally, RNA was isolated from gb-MSCs, and miRNA modifications were analyzed using the RAffymetrix miRNA microarray Results: We showed that glioblastoma-conditioned medium increased gb-MSC proliferation and migration and was capable of inducing gb-MSC differentiation into pericytes. Glioblastoma secreted angiogenic factors and gb-MSCs incubated in malignant glioblastoma-conditioned medium formed more tube-like structures, and these cells also adhered to tube-like vessels formed by human umbilical vein endothelial cells (HUVECs) on Matrigel to maintain tumor vascular structure in vitro. miRNA expression were also modified in gb-MSCs cultured in malignant glioblastoma-conditioned medium in vitro. Conclusion: These results provide new insight into the functional effects of a subpopulation of MSCs in glioblastoma and may help in the development of novel therapies for solid tumors.


PLoS ONE ◽  
2014 ◽  
Vol 9 (11) ◽  
pp. e113572 ◽  
Author(s):  
Guo-hu Di ◽  
Yang Liu ◽  
Ying Lu ◽  
Jin Liu ◽  
Chutse Wu ◽  
...  

2021 ◽  
Vol 30 ◽  
pp. 096368972199377
Author(s):  
Rui Zhang ◽  
Qiaoxia Zhang ◽  
Zhiyu Zou ◽  
Zheng Li ◽  
Meng Jin ◽  
...  

Mesenchymal stem cells derived from bone marrows (BMSCs) and curcumin derived from turmeric were used for osteoarthritis (OA) treatment, respectively. We invested the effects of curcumin supplementation for BMSC therapeutic effects. In vitro, rat BMSCs were identified by dual-immunofluorescent staining of CD44 and CD90, and flow cytometry. Primary articular chondrocytes were identified by toluidine blue staining and immunofluorescent staining of Col2a1. EdU incorporation, migration assay, real-time quantitative polymerase chain reaction, and Western blot analyses were performed to evaluate the alterations of chondrocytes cocultured with BMSCs. In vivo, the rat model of OA was established by monoiodoacetic acid. After intra-articular injection of allogeneic BMSCs, articular cartilage damage and OA progression were evaluated by histological staining, and Osteoarthritis Research Society International and Mankin score evaluation. Although curcumin alone did not improve cell viability of primary articular chondrocytes, it promoted proliferation and migration of chondrocytes when cocultured with BMSCs. Meanwhile, the expression of anabolic genes in chondrocytes was remarkably increased both at mRNA and protein levels. In OA rats, curcumin and BMSCs cooperated to greatly promote articular cartilage repair and retard OA progression. Therefore, curcumin supplementation enhanced the BMSC function for the proliferation and migration of articular chondrocytes, and anabolic gene expression of extracellular matrix in articular chondrocytes in vitro, and the generation of articular cartilage in vivo. Our study shed light on the potential clinical application of curcumin cooperated with BMSCs in cartilage repair for OA treatment.


2021 ◽  
Author(s):  
Pengdong Li ◽  
Shuang Lv ◽  
Wenyue Jiang ◽  
Lihui Si ◽  
Baojian Liao ◽  
...  

Abstract BackgroundOsteoarthritis (OA) is one of the most common joint diseases and a major public health concern. Current therapies for OA can relieve symptoms but offer no potential for cartilage regeneration. Mesenchymal stem cells (MSCs) have been widely used for the treatment of OA owing to their paracrine secretion of trophic factors, a phenomenon in which exosomes may play a major role. Here, we investigated the potential of exosomes from human umbilical cord-derived MSCs (hUC-MSCs-Exos) at alleviating OA.MethodshUC-MSCs were isolated, cultured, and identified based on the expression of MSC markers and multipotency differentiation. hUC-MSCs-Exos were harvested from hUC-MSC conditioned medium using a sequential centrifugation method. Transmission electron microscopy, dynamic light scattering, flow cytometry, and western blotting were used to identify the exosomes. The effects of hUC-MSCs-Exos on the proliferation and migration of human chondrocytes were evaluated using the cell counting kit-8, EdU-555 cell proliferation kit, and transwell assays. Annexin V-FITC/PI staining and flow cytometry were used to evaluate the effect of exosomes on chondrocyte apoptosis. An in vitro model of human articular chondrocytes treated with interleukin 1 beta (IL-1β) was used to evaluate the effects of exosomes; analyses involved using quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence, and western blotting. The role of exosomes in macrophage polarization was examined in the monocyte cell line, THP-1. Rats with surgically induced OA (ACLT+pMMx method) were intra-articularly injected with hUC-MSCs-Exos. The efficacy of exosome injections was assessed using hematoxylin and eosin and safranin-O and fast green staining, and immunohistochemistry.ResultsWe confirmed the superior efficacy of hUC-MSCs-Exos at promoting chondrocyte proliferation and migration and inhibiting chondrocyte apoptosis. Additionally, hUC-MSCs-Exos reversed IL-1β-induced injury in vitro. hUC-MSCs-Exos could inhibit the secretion of pro-inflammatory factors, promote the expression of anti-inflammatory factors, and regulate the polarization of macrophages. hUC-MSCs-Exos attenuated the progression of OA and prevented severe damage to the knee articular cartilage in the rat OA model. ConclusionshUC-MSCs-Exos exerted immunomodulatory and therapeutic effects in a rat model of OA. These exosomes derived from hUC-MSCs can potentially serve as treatments for OA.


Sign in / Sign up

Export Citation Format

Share Document