PDTC inhibits apoptosis and phenotypic transformation of co-culture of myeloma cells and renal tubular epithelial cells by reducing the secretion of light chain protein

2020 ◽  
Author(s):  
Xiaoyan Yu ◽  
Jie Bao ◽  
Xinyu Cui ◽  
Fengxia Du ◽  
Yuefei Wang

Abstract Background The human myeloma cell line RPMI-8226 is co-cultured with human proximal tubular epithelial cell HK-2, which is commonly used to simulate the renal environment in patients with multiple myeloma. This study aimed to investigate the effects of NFκB inhibitor PDTC on the viability, apoptosis and cell phenotype of HK-2 cells in the co-culture system of myeloma cells in renal tubular epithelial cells.Methods An in vivo environment was simulated through a cell co-culture system. RPMI-8226 cells and HK-2 cells were inoculated in the co-culture chamber and cultured for 24 h to establish the co-culture system. PDTC was used in the single culture group and the co-culture group, respectively. The activity of HK-2 cells and RPMI-8226 cells in each group was detected by MTT. An immunoturbidimetric assay was performed to assess the effect of PDTC on secretion of κ light chain and λ light chain in RPMI-8226 cells. Flow cytometry was used to detect apoptosis of HK-2 cells. Western blot was carried out to detect NF-κB activation in RPMI-8226 myeloma cells, as well as the expression levels of caspase-3, bcl-2, Bax, E-cadherin, and α-SMAin HK-2 cells. Caspse-3 assay kit was used to detect the activity of caspase-3. The effect of PDTC on the secretion of κ light chain and λ light chain of RPMI-8226 cells was detected by immunoturbidimetry and the ratio was calculated.Results PDTC had a potent inhibitory effect on proliferation of RPMI-8226 cells in a dose- and time-dependent manner. PDTC had no significant effect on the viability of HK-2 cells cultured alone, and the addition of PDTC to the co-culture system significantly increased the viability of HK-2 cells. PDTC did not significantly change the apoptosis of HK-2 cells cultured alone, but could reduce the apoptosis of renal tubular epithelial cells by regulating the activity of caspase3 and the ratio of bcl2/bax in HK-2 cells in the co-culture system. After PDTC treatment, the expression of cell surface marker E-cadherin decreased, and the expression of α-SMA increased, which induced the renal interstitial fibrosis. The secretion of κ light chain and λ light chain of RPMI-8226 cells was significantly decreased after the addition of PDTC, but the ratio was not changed.Conclusions PDTC can inhibit the cell activity, promote apoptosis, and reduce the secretion of secretion of κ light chain and λ light chain through inhibiting the NF-κB pathway activation of myeloma cell RPMI-8226, leading to increased activity of renal tubular epithelial cells HK-2 in the co-culture system, decreased apoptosis, and renal interstitial fibrosis.

Author(s):  
Xiaoyan YU ◽  
Jie BAO ◽  
Xinyu CUI ◽  
Fengxia DU ◽  
Yuefei WANG ◽  
...  

Background: We investigate the effects of NFκB inhibitor pyrrolidinedithiocarbamic acid ammonium salt (PDTC) on the viability, apoptosis and cell phenotype of HK-2 cells in the co-culture system of myeloma cells in renal tubular epithelial cells. Methods: This study was performed in Qiqihar Medical University, Qiqihar, China from Jun 2018 to Jan 2019. RPMI-8226 cells and HK-2 cells were inoculated in the co-culture chamber and cultured to establish the co-culture system. An immunoturbidimetric assay was performed to detect κ light chain and λ light chain in RPMI-8226 cells. The effect of PDTC on the secretion of κ light chain and λ light chain of RPMI-8226 cells was detected by immunoturbidimetry and the ratio was calculated. Results: PDTC significantly increased the viability of HK-2 cells. PDTC reduced the apoptosis of renal tubular epithelial cells. After PDTC treatment, the expression of cell surface marker E-cadherin decreased, and the expression of α-SMA increased, which induced the renal interstitial fibrosis. The secretion of κ light chain and λ light chain of RPMI-8226 cells was significantly decreased after the addition of PDTC, but the ratio was not changed. Conclusion: PDTC can inhibit the cell activity, promote apoptosis, and reduce the secretion of secretion of κ light chain and λ light chain through inhibiting the NF-κB pathway activation of myeloma cell RPMI-8226.


2011 ◽  
Vol 30 (7-8) ◽  
pp. 396-403 ◽  
Author(s):  
Maria Fragiadaki ◽  
Abigail S. Witherden ◽  
Tomoyo Kaneko ◽  
Sonali Sonnylal ◽  
Charles D. Pusey ◽  
...  

2017 ◽  
Vol 46 (4) ◽  
pp. 333-342 ◽  
Author(s):  
Huifang Liu ◽  
Jiachuan Xiong ◽  
Ting He ◽  
Tangli Xiao ◽  
Yan Li ◽  
...  

Background: Hyperuricemia is an independent risk factor for causing chronic kidney disease and contributes to kidney fibrosis. After urate crystals get deposited in the kidney, they can cause hyperuricemia nephropathy, leading to glomerular hypertrophy and renal tubular interstitial fibrosis. Recent data showed that uric acid (UA) could induce epithelial mesenchymal transition (EMT) of renal tubular cells, in which NRLP3 inflammatory pathway was involved. However, whether TLR4/NF-κB signaling pathway is also involved in EMT of renal tubular cells induced by UA is not clear. Methods: Human renal tubular epithelial cells (HK-2) were directly treated with UA and the phenotypic transition was detected by morphological changes and the molecular markers of EMT. The activation of the TLR4/NF-κB signaling pathway induced by UA was measured by Western blot and its involvement was further confirmed by the inhibition of NF-κB activation or knockdown of toll like receptor 4 (TLR4) expression. Results: UA induced obvious morphological changes of HK-2 cell, accompanied with altered molecular markers of EMT including fibronectin, α-SMA and E-cadherin. In addition, UA significantly upregulated the gene expression of interleukin-1β and tumor necrosis factor-α in a time- and dose-dependent manner. Furthermore, UA significantly activated the TLR4/NF-κB signaling pathway in HK-2 cells, while the inhibition of the TLR4 expression by siRNA and NF-κB activation by PDTC significantly attenuated EMT induced by UA in HK-2 cells. Conclusions: UA can induce EMT in renal tubular epithelial cells by the activation of the TLR4/NF-κB signaling pathway, and the targeted intervention of the TLR4/NF-κB signaling pathway might effectively inhibit UA-induced renal interstitial fibrosis mediated by EMT.


2021 ◽  
Vol 21 (2) ◽  
pp. 1266-1271
Author(s):  
Ping Zhao ◽  
Ting Li ◽  
Zhi Li ◽  
Lei Cao ◽  
Youliang Wang ◽  
...  

Gold nanoparticles (GNPs) are widely used in life sciences and medicine due to their simple preparation, stable physical and chemical properties, controllable optical properties and no significant toxicity. However, in recent years, studies have found that there are still many uncertain factors in the application of gold nanoparticles in the field of biomedicine, and there are few studies on the main excretion organs and kidneys of the body, especially the toxicological effects under the disease state have not been reported. Obviously, carrying out relevant research is of great significance for accelerating the clinical application of GNPs. Chronic kidney disease (CKD) is a group of chronic progressive diseases that have high prevalence and high mortality and are serious threats to human life and health. Renal tubular injury and interstitial fibrosis are key factors in renal dysfunction in chronic kidney disease. Drug and toxic kidney damage mostly involve renal tubular epithelial cells; hypoxia is the most common pathological condition of cells. In renal lesions, renal tubular epithelial cells often have hypoxia. Based on this, we propose the hypothesis of this study: glomerular filtration membrane damage in kidney disease, GNPs increase in urine, followed by reabsorption of renal tubular epithelial cells, thereby causing damage to the latter; if accompanied by hypoxia, GNPs it will aggravate renal tubular epithelial cell damage and promote tubulointerstitial fibrosis. In order to verify the above hypothesis, this study used a mouse model of adriamycin nephropathy and tubular epithelial cells and macrophages in vitro, and observed the damage of GNPs on renal tubular epithelial cells by various means, and explored related mechanisms. The results show that under normal oxygen conditions, GNPs can induce autophagy after cell entry, which can damage damaged proteins and organelles to maintain cell survival. In the absence of oxygen, nanoparticles entering cells increase and induce excessive autophagy. In the absence of oxygen, GNPs also aggregate in macrophages, which can cause decreased cell proliferation activity and induce activation of macrophage inflammasome, which induces inflammatory response: GNPs-induced secretion of hypoxic macrophages can be promoted.


Sign in / Sign up

Export Citation Format

Share Document