saos2 cell
Recently Published Documents


TOTAL DOCUMENTS

3
(FIVE YEARS 2)

H-INDEX

1
(FIVE YEARS 0)

Author(s):  
BiBi Fatemeh Haghir Alsadat ◽  
Zeinab Amirpour ◽  
Behrad Nafisi

Background: Creating a new berberine liposome with high encapsulation efficiency and slow release formulation in the treatment of cancer is a new issue. Therefore, the aim of current study was to develop slow release berberine-containing nanoliposome for delivery to bone cancer cells Saos2. Materials and Methods: In this experimental study, after synthesis nanoliposomal formulation, physical parameters, including size, zeta potential, and drug loading, in liposome were assessed using different techniques. Saos2 cell line was incubated in micro-plates containing Dulbecco's Modified Eagle's medium (DMEM) and FBS at 37˚C and 5% CO2. Cytotoxicity of nanoliposome was assessed using MTT assay. The release of drug from nanoliposome was assessed by dialysis method. P< 0.05 was assumed significant. Results: The size of drug-free nanoliposome and drug nanoliposome (berberine-nanoliposome) was 112.1 and 114.9 nm, respectively. The zeta potential of drug-free nanoliposome and drug- nanoliposome was –16.1 and –1.9 mv, respectively. There was no significant difference between control and drug-free nanoliposome groups regarding viability (p>0.05). The viability of cells in different concentration of nanoliposome containing berberines in Saos2 cell line was significantly higher than that in free berberines (p<0.05). The release of berberine at temperature 37 º C and pH 7.4 showed that approximately 47% of the drug was released in the first 12 hours of study and then the slow release of drug was continued. IC50 value of free berberine and berberine containing nanoliposome was 137.3 and 52.2 µg / ml, respectively. Conclusion: According to these findings, IC50 value of free berberine was 2.67 times more than berberine containing nanoliposome, indicating that nanoliposome containing berberine had more inhibition on growth of cancer cells than free berberine. In addition, the drug release was slow exposing the drug to the tumor for longer time at a lower dose and fewer injections, increasing the effect of the drug on cancer cells.


2020 ◽  
Vol 13 (8) ◽  
Author(s):  
Behnoosh Tahbazlahafi ◽  
Malihe Paknejad ◽  
Shiva Shahmohamadnejad ◽  
Alireza Mirzaei ◽  
Khodamorad Jamshidi ◽  
...  

Background: Osteosarcoma (OS) is the most common type of bone malignancy. Many studies have attempted to find the association between microRNAs and cancer-associated processes. Alterations in miRNA expression through genetic or epigenetic changes, impairment of transcription factors, and ectopic expression of miRNAs induce the development and progression of cancer. Although miR-135b has been thoroughly documented as an oncogene in the majority of studies, some controversies remain about the conflicting role of miR-135b as a tumor-suppressor. Objectives: The present study aimed at investigating the oncogenic and/or tumor-suppressing role of miR-135b in human OS. Methods: In this study, 21 OS tissue samples, along with 21 adjacent bone tissues (normal) as control specimens were collected to analyze the expression of miR-135b. The Saos2 cell-line was transiently transfected with the miR-135b mimic and inhibitor to assess its effect on two critical transcription factors, namely FOXO-1 and c-Myc. qRT-PCR was performed to quantify the expression of miR-135b in both OS tissues and the Saos2 cell-line. The MTT, cell migration, and cell invasion assays were used to characterize the miR-135b function. The western blot analysis was carried out to monitor the targets of miR-135b. Finally, the changes in cellular functions such as migration and invasion, following the transfection of miR-135b mimic and inhibitor, were verified. Results: The results showed that in comparison with the adjacent normal bone tissues, the expression of miR-135b was higher in OS tissue samples, which inversely correlated with the expression rate of FOXO-1, whereas the expression of c-Myc had a direct relationship to miR-135b expression. Functionally, the miR-135b mimic led to an increase in cell proliferation, invasion, and migration of OS cancer cells. Conclusions: MiR-135b induces the proliferation and invasion of OS cells by the degradation of FOXO-1 and upregulation of c-Myc.


Sign in / Sign up

Export Citation Format

Share Document