Silencing PEX26 as an Unconventional Mode to Kill Drug-Resistant Cancer Cells and Forestall Drug Resistance

Autophagy ◽  
2021 ◽  
Author(s):  
Michael S. Dahabieh ◽  
Fan Huang ◽  
Christophe Goncalves ◽  
Raúl Ernesto Flores González ◽  
Sathyen Prabhu ◽  
...  
2020 ◽  
Author(s):  
Jinyan Zhao ◽  
Weilan Lan ◽  
Jun Peng ◽  
Bin Guan ◽  
Jie Liu ◽  
...  

Abstract Background: Multidrug resistance (MDR) is a critical reason of cancer chemotherapy failure. Babao dan (BBD) is a classical and famous traditional Chinese patent medicine, which has been reported to has anti-gastric cancer activity. However, the roles and molecular mechanisms of the reversal of MDR of gastric cancer by BBD have not been well described until now. Methods: SGC-7901 and SGC-7901/DDP cells were used in this study, and drug resistance and evaluation of the reversal effect of BBD was determined using MTT assays in SGC7901/DDP cells. Doxorubicin (DOX) and Rhodamin123 (Rho123) staining was performed to assess BBD effects on drug accumulation and efflux of drug-resistant gastric cancer cells. Cell apoptosis was directly assessed using DAPI staining. Apoptotic and dead cells were detected by flow cytometry after staining with Annexin V-FITC and propidium iodide (PI). Cyto-ID assays were performed to examine cellular autophagy. Changes in cell protein expression of ABCB1, ABCC1, ABCG2, Bax, Bcl-2, caspase-3, cleaved-caspase-3, LC3, p62, Beclin1 and the PI3K/AKT/mTOR pathway were detected by Western blot. Inhibition of autophagy with 3-MA, chloroquine (CQ) and PI3K antagonist (LY294002) or agonist (740Y-P) , uncovered a role for the potentially downregulated signaling pathway, PI3K/AKT/mTOR.Results: The SGC7901/DDP cell line exhibited multi-drug resistance to DDP, DOX and 5-fluorouracil (5-FU) and the drug resistant index (RI) of DDP, DOX and 5-FU were 1.86, 1.50 and 47.70, respectively. BBD reversed the MDR of SGC7901/DDP cells by increasingDOX accumulation, reducing Rh123 efflux and down-regulating the expression of ABCB1, ABCC1, ABCG2. Furthermore, BBD induced apoptosis in SGC7901/DDP cells through regulating caspase-3, cleaved-caspase-3, Bax and Bcl-2. Moreover, BBD induced autophagy in DDP-resistant gastric cancer cells via regulating p62, LC3 and Beclin1. Pathway analyses suggested BBD may inhibit PI3K/AKT/mTOR pathway activity and subsequent autophagy induction. Conclusions: BBD may reverse the MDR of gastric cancer cells, and promote autophagic death via inactivation of the PI3K/AKT/mTOR signaling pathway.


Metabolites ◽  
2020 ◽  
Vol 10 (7) ◽  
pp. 289 ◽  
Author(s):  
Xun Chen ◽  
Shangwu Chen ◽  
Dongsheng Yu

Metabolic reprogramming is one of the hallmarks of tumors. Alterations of cellular metabolism not only contribute to tumor development, but also mediate the resistance of tumor cells to antitumor drugs. The metabolic response of tumor cells to various chemotherapy drugs can be analyzed by metabolomics. Although cancer cells have experienced metabolic reprogramming, the metabolism of drug resistant cancer cells has been further modified. Metabolic adaptations of drug resistant cells to chemotherapeutics involve redox, lipid metabolism, bioenergetics, glycolysis, polyamine synthesis and so on. The proposed metabolic mechanisms of drug resistance include the increase of glucose and glutamine demand, active pathways of glutaminolysis and glycolysis, promotion of NADPH from the pentose phosphate pathway, adaptive mitochondrial reprogramming, activation of fatty acid oxidation, and up-regulation of ornithine decarboxylase for polyamine production. Several genes are associated with metabolic reprogramming and drug resistance. Intervening regulatory points described above or targeting key genes in several important metabolic pathways may restore cell sensitivity to chemotherapy. This paper reviews the metabolic changes of tumor cells during the development of chemoresistance and discusses the potential of reversing chemoresistance by metabolic regulation.


2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Yaoyue Qi ◽  
Weiwei Qi ◽  
Shihai Liu ◽  
Libin Sun ◽  
Aiping Ding ◽  
...  

Abstract Background The issue of drug resistance in gastric cancer has attracted global attention. TSPAN9, a 4-transmembrane protein that plays an important role in tumor progression and signal transduction, has been found to be closely related to tumor invasion, metastasis, and autophagy. Methods Immunoblotting was used to evaluate TSPAN9 expression in parental and drug-resistant gastric cancer cells. Functional assays, such as the CCK-8 assay, were used to detect the proliferation of gastric cancer cells and the response of TSPAN9 to 5-fluorouracil (5-FU). Western blotting was used to analyze the expression of constituents of the PI3K/AKT/mTOR-mediated autophagy pathway induced by TSPAN9. Coimmunoprecipitation was performed to assess the specific mechanism by which TSPAN9 affects the PI3K pathway. Results We demonstrated that TSPAN9 is overexpressed in 5-FU-resistant cells compared to parental cells. 5-FU-mediated inhibition of cell proliferation can be significantly restored by increasing TSPAN9 expression, and inhibiting this expression in drug-resistant cells can restore the sensitivity of the cells to 5-FU. In addition, TSPAN9 also significantly promoted autophagy in gastric cancer cells in vitro. Further studies indicated that TSPAN9 downregulates the expression of PI3K and proteins associated with PI3K-mediated autophagy. In addition, TSPAN9 interacts with PI3K and inhibits its catalytic activity. Conclusion The current study reveals the important role of TSPAN9 in drug resistance to 5-FU in gastric cancer. It also provides a new target to clinically address drug-resistant gastric cancer and will contribute to the treatment strategy of this disease.


2020 ◽  
Vol 41 (10) ◽  
pp. 1329-1340 ◽  
Author(s):  
Ga-Bin Park ◽  
Jee-Yeong Jeong ◽  
Daejin Kim

Abstract In cancer, resistance to chemotherapy is one of the main reasons for therapeutic failure. Cells that survive after treatment with anticancer drugs undergo various changes, including in cell metabolism. In this study, we investigated the effects of AKT-mediated miR-125b-5p alteration on metabolic changes and examined how these molecules enhance migration and induce drug resistance in colon cancer cells. AKT1 and AKT3 activation in drug-resistant colon cancer cells caused aberrant downregulation of miR-125b-5p, leading to GLUT5 expression. Targeted inhibition of AKT1 and AKT3 restored miR-125b-5p expression and prevented glycolysis- and lipogenesis-related enzyme activation. In addition, restoring the level of miR-125b-5p by transfection with the mimic sequence not only significantly blocked the production of lactate and intracellular fatty acids but also suppressed the migration and invasion of chemoresistant colon cancer cells. GLUT5 silencing with small interfering RNA attenuated mesenchymal marker expression and migratory activity in drug-resistant colon cancer cells. Additionally, treatment with 2,5-anhydro-d-mannitol resensitized chemoresistant cancer cells to oxaliplatin and 5-fluorouracil. In conclusion, our findings suggest that changes in miR-125b-5p and GLUT5 expression after chemotherapy can serve as a new marker to indicate metabolic change-induced migration and drug resistance development.


2019 ◽  
Author(s):  
Shifeng Shi ◽  
Xin Huang ◽  
Xiao Ma ◽  
Xiaoyan Zhu ◽  
Qinxian Zhang

AbstractPurposeChemotherapy resistance of esophageal cancer is a key factor affecting the postoperative treatment of esophageal cancer. Among the media that transmit signals between cells, the exosomes secreted by tumor cells mediate information transmission between tumor cells, which can make sensitive cells obtain resistance. Although some cellular exosomes play an important role in tumor’s acquired drug resistance, the related action mechanism is still not explored specifically.MethodsTo elucidate this process, we constructed a cisplatin-resistant esophageal cancer cell line, and proved that exosomes conferring cellular resistance in esophageal cancer can promote cisplatin resistance in sensitive cells. Through high-throughput sequencing analysis of the exosome and of cells after stimulation by exosomes, we determined that the miRNA193 in exosomes conferring cellular resistance played a key role in sensitive cells acquiring resistance to cisplatin. In vitro experiments showed that miRNA193 can regulate the cell cycle of esophageal cancer cells and inhibit apoptosis, so that sensitive cells can acquire resistance to cisplatin. An in vivo experiment proved that miRNA193 can promote tumor proliferation through the exosomes, and provide sensitive cells with slight resistance to cisplatin.ResultsSmall RNA sequencing of exosomes showed that exosomes in drug-resistant cells have 189 up-regulated and 304 down-regulated miRNAs; transcriptome results showed that drug-resistant cells treated with drug-resistant cellular exosomes have 3446 high-expression and 1709 low-expression genes; correlation analysis showed that drug-resistant cellular exosomes mainly affect the drug resistance of sensitive cells through paths such as cytokine–cytokine receptor interaction, and the VEGF and Jak-STAT signaling pathways; miRNA193, one of the high-expression miRNAs in drug-resistant cellular exosomes, can promote drug resistance by removing cisplatin’s inhibition of the cell cycle of sensitive cells.ConclusionSensitive cells can become resistant to cisplatin through acquired drug-resistant cellular exosomes, and miRNA193 can make tumor cells acquire cisplatin resistance by regulating the cell cycle.


Author(s):  
Jiaqi Xiao ◽  
Meixiang Gao ◽  
Qiang Diao ◽  
Feng Gao

: Drug resistance including multidrug resistance resulting from different defensive mechanisms in cancer cells is the leading cause of the failure about the cancer therapy, making it an urgent need to develop more effective anticancer agents. Chalcones, widely distributed in nature, could act on diverse enzymes and receptors in cancer cells. Accordingly, chalcone derivatives possess potential activity against various cancers including drug-resistant even multidrug-resistant cancer. This review outlines the recent development of chalcone derivatives with potential activity against drug-resistant cancers covering articles published between 2010 and 2020, so as to facilitate further rational design of more effective candidate.


Cancers ◽  
2018 ◽  
Vol 10 (12) ◽  
pp. 483 ◽  
Author(s):  
Meghan Leary ◽  
Sarah Heerboth ◽  
Karolina Lapinska ◽  
Sibaji Sarkar

Cancer drug resistance is an enormous problem. It is responsible for most relapses in cancer patients following apparent remission after successful therapy. Understanding cancer relapse requires an understanding of the processes underlying cancer drug resistance. This article discusses the causes of cancer drug resistance, the current combination therapies, and the problems with the combination therapies. The rational design of combination therapy is warranted to improve the efficacy. These processes must be addressed by finding ways to sensitize the drug-resistant cancers cells to chemotherapy, and to prevent formation of drug resistant cancer cells. It is also necessary to prevent the formation of cancer progenitor cells by epigenetic mechanisms, as cancer progenitor cells are insensitive to standard therapies. In this article, we emphasize the role for the rational development of combination therapy, including epigenetic drugs, in achieving these goals.


Author(s):  
GA-BIN PARK ◽  
Daejin Kim

Abstract Background CD248, also called endosialin or tumor endothelial marker-1 (TEM1), is markedly upregulated in almost all cancers, including colon cancers. Changes in microRNA (miRNA) profiles are one of the direct causes of cancer development and progression. In this study, we investigated whether a change in CD248 expression in colon cancer cells could induce drug resistance after chemotherapy, and we explored the relationship between miR-125b-5p levels and CD248 expression in Toll-like receptor (TLR)-modified chemoresistant colon cancer cells. Methods We identified the one of the downregulated miRNAs in drug-resistant HCT8 cells using Affymetrix Genechip miRNA 4.0 array process and validated the expressional change of chemoresistant HCT-116 and HT-29 cells. Interaction between Sp1 and miR-125b-5p was confirmed by miScript target protector assay. To characterize the underlying mechanisms involved in CD248 expression, we adapted the several biological analysis techniques, including RNA-binding Protein Immunoprecipitation (RIP), migration analysis, real-Time PCR, western blot analysis, and gene silencing using siRNA. Results TLR2/6 and TLR5 upregulation in drug-resistant colon cancer cells contributed to miR-125b-5p downregulation and Sp1-mediated CD248 upregulation via NF-κB activation. Exposure to specific TLR2/6 or TLR5 ligands enhanced the expression of mesenchymal markers as well as the migratory activity of oxaliplatin (Ox)- or 5-fluorouracil (5-Fu)-resistant colon cancer cells. The transfection of a synthetic miR-125b-5p mimic into chemoresistant cells prevented Sp1 and CD248 activation and significantly impaired invasive activity. Furthermore, Sp1 or CD248 gene silencing as well as miR-125b-5p overexpression markedly reversed drug resistance and inhibited epithelial-mesenchymal transition (EMT) in colon cancer cells. Conclusions Taken together, these results suggest that changes in miR-125b-5p levels play an important role in Sp1-mediated CD248 expression and the development of drug resistance in TLR-mutated colon cancer cells.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 756-756
Author(s):  
Jayakumar R Nair ◽  
Louise M Carlson ◽  
Kelvin P Lee

Abstract Despite novel chemotherapeutic regimens, myeloma patients invariably develop drug resistance and eventually die due to disease relapse, highlighting the necessity for newer therapies to extend survival. Interactions with bone marrow stroma are essential for drug resistance and survival in myeloma (MM). Not much is known about the specific cell types or molecular interactions that mediate this. Clinical studies have shown that expression of CD28 (the prototypic T cell costimulatory molecule) on MM correlates with disease progression. Our recent publication and abstracts (ASH2012 #722, ASH2011 #147, ASH2010 #132) show that interaction with dendritic cells (DC) in the bone marrow provide pro-survival signals to MM through CD28 via interactions with its ligands CD80/CD86 on DC or CD86 on other MM. Clinical studies have separately shown that CD28 or CD86 expression on MM portends poor survival in myeloma patients. In our previous ASH abstracts, we have shown that blocking MM CD28 interactions with CD80/CD86 on DC or other MM using novel reagents such as CTLA4Ig (Abatacept®, a recombinant fusion protein between CTLA4 and human IgFc) or blocking αCD28(Fab) fragments can reduce CD28 mediated survival in MM. In contrast, activating CD28 on MM with agonistic antibodies improves survival against different therapeutics or serum starvation. Another mechanism by which cancer cells survive exogenous stress is via redox regulation and our data that show that BSA (a known scavenger of reactive oxygen species (ROS)) can alleviate CTLA4Ig mediated sensitization of MM cells (Fig 1) suggest involvement of redox regulation in CD28 mediated MM survival. Intrinsic oxidative stress is a hallmark of cancer and is associated with abnormal cancer growth and progression. Others have reported that cancer cells adapt to intrinsic oxidative stress by developing enhanced anti-oxidant capacity, and are more resistant to exogenous stress. Literature also suggests that cancer cells that have higher intrinsic oxidative stress are also more likely to be sensitive to any disruption of redox regulation than normal cells. In MM, flow cytometric analysis using ROS dye CM-DCFDA show significantly higher (8-9 fold) basal ROS levels in the drug resistant U266 as opposed to the drug sensitive cell line MM.1S (Fig 2). Thioredoxin (TRX1) is a key ROS induced anti-oxidant protein essential for redox regulation and survival in many types of cancers. But not much is known about its role in myeloma survival. Interestingly, gene expression analysis of public datasets of plasma cells from normal, MGUS and myeloma patients show significant increases in the levels of TRX1 with disease progression. Further, within the myeloma patient group, TRX1 levels were significantly higher in the “relapsed” group relative to new or smoldering MM groups (Fig 3). In contrast, the levels of thioredoxin-interacting protein (TXNIP), a negative regulator of thioredoxin was significantly lower in myeloma patients and in the relapsed group, compared to normal or new diagnosed patients respectively (Fig 3). Expression trends for TRX1 and TXNIP were inversely correlated across patient groups. While CD28 activation in MM can alleviate drug induced cell death in myeloma, it could not overcome cell death induced by the specific TRX1 inhibitor PX-12 (Biomira Inc, currently under Phase 2 clinical trials for pancreatic cancer). Moreover, viability assays show that high-ROS cell line U266 was 4 fold more sensitive to PX-12 than the low-ROS MM.1S cells which is interesting since U266 is drug resistant and is thus representative of relapsed myeloma. ROS assays with PX-12 on MM show rapid dose dependent increases in ROS levels in U266 cells, while it was much lower in MM.1S suggesting a rationale for the higher sensitivity of U266. This was reversed when a ROS scavenger N-acetyl cysteine (NAC) was added. NAC also completely abrogated PX-12 mediated apoptosis in both myeloma cell lines U266 and MM.1S suggesting that PX-12's activity was solely via disruption of redox regulation. Our data reveal an important redox regulatory mechanism mediated by thioredoxin which play a supportive role for CD28 mediated survival in myeloma and disease relapse, disruption of which could selectively target relapsed drug resistant myeloma. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document