2015
DOI: 10.1007/s10585-015-9731-4
|View full text |Cite
|
Sign up to set email alerts
|

Eight proteins play critical roles in RCC with bone metastasis via mitochondrial dysfunction

Abstract: Most kidney cancers are renal cell carcinomas (RCC). RCC lacks early warning signs and 70 % of patients with RCC develop metastases. Among them, 50 % of patients having skeletal metastases developed a dismal survival of less than 10 % at 5 years. Therefore, exploring the key driving proteins and pathways involved in RCC bone metastasis could benefit patients’ therapy and prolong their survival. We examined the difference between the OS-RC-2 cells and the OS-RC-2-BM5 cells (subpopulation from OS-RC-2) of RCC wi… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
2

Citation Types

0
22
0

Year Published

2016
2016
2024
2024

Publication Types

Select...
6
1

Relationship

1
6

Authors

Journals

citations
Cited by 17 publications
(22 citation statements)
references
References 50 publications
(35 reference statements)
0
22
0
Order By: Relevance
“…In our previous proteome analysis of the total cellular protein from RCC tumor cells, STIP1 protein was found to be 4-fold more abundant in the bone-seeking cells than in the control parental cells [12], and STIP1 was observed to be translocated to the cell surface and secreted out of cells [13, 14], which may mediate extracellular tumor-niche interactions. To further define the subcellular location of the increased STIP1 protein, and study whether tumor cells actively secrete STIP1 protein, we conducted two experiments and found First, After culturing RCC tumor cells were cultured in serum-free medium for 24 h, we observed that the culture media clearly contained STIP1 but no intracellular GAPDH protein (Figure 1A).…”
Section: Resultsmentioning
confidence: 99%
See 3 more Smart Citations
“…In our previous proteome analysis of the total cellular protein from RCC tumor cells, STIP1 protein was found to be 4-fold more abundant in the bone-seeking cells than in the control parental cells [12], and STIP1 was observed to be translocated to the cell surface and secreted out of cells [13, 14], which may mediate extracellular tumor-niche interactions. To further define the subcellular location of the increased STIP1 protein, and study whether tumor cells actively secrete STIP1 protein, we conducted two experiments and found First, After culturing RCC tumor cells were cultured in serum-free medium for 24 h, we observed that the culture media clearly contained STIP1 but no intracellular GAPDH protein (Figure 1A).…”
Section: Resultsmentioning
confidence: 99%
“…Under the normal monolayer culture condition, both MTT assay and flow cytometry cell cycle analysis revealed that the bone-seeking cells proliferated faster than the parental cells [11, 12]. To test whether STIP1 modulates the proliferation of tumor cells, we added human recombinant STIP1 (hrSTIP1) to the culture media.…”
Section: Resultsmentioning
confidence: 99%
See 2 more Smart Citations
“…This inhibition was most likely accomplished by masking the histone lysines from acetylation, consequently silencing HATdependent transcription [16]. The knockdown of SET inhibits cell migration and invasion by increasing the activity and expression of PP2Ac and decreasing the expression of matrix metalloproteinase 9 (MMP-9) [17]; low levels of SET expression are associated with bone metastasis in renal cell carcinoma [18]. Therefore, using a bioinformatics prediction, we found that SET was a potential target of miR-125b; therefore, we sought to assess the expression of miRNA, its effect in human EOC, and its association with SET expression and epithelial-mesenchymal transition (EMT) in human EOC.…”
Section: Introductionmentioning
confidence: 99%