scholarly journals Modified Gold Nanoparticles for Efficient Delivery of Betulinic Acid to Cancer Cell Mitochondria

2021 ◽  
Vol 22 (10) ◽  
pp. 5072
Author(s):  
Olakunle Oladimeji ◽  
Jude Akinyelu ◽  
Aliscia Daniels ◽  
Moganavelli Singh

Advances in nanomedicine have seen the adaptation of nanoparticles (NPs) for subcellular delivery for enhanced therapeutic impact and reduced side effects. The pivotal role of the mitochondria in apoptosis and their potential as a target in cancers enables selective induction of cancer cell death. In this study, we examined the mitochondrial targeted delivery of betulinic acid (BA) by the mitochondriotropic TPP+-functionalized epigallocatechin gallate (EGCG)-capped gold NPs (AuNPs), comparing the impact of polyethylene glycol (PEG) and poly-L-lysine-graft-polyethylene glycol (PLL-g-PEG) copolymer on delivery efficacy. This included the assessment of their cellular uptake, mitochondrial localization and efficacy as therapeutic delivery platforms for BA in the human Caco-2, HeLa and MCF-7 cancer cell lines. These mitochondrial-targeted nanocomplexes demonstrated significant inhibition of cancer cell growth, with targeted nanocomplexes recording IC50 values in the range of 3.12–13.2 µM compared to that of the free BA (9.74–36.31 µM) in vitro, demonstrating the merit of mitochondrial targeting. Their mechanisms of action implicated high amplitude mitochondrial depolarization, caspases 3/7 activation, with an associated arrest at the G0/G1 phase of the cell cycle. This nano-delivery system is a potentially viable platform for mitochondrial-targeted delivery of BA and highlights mitochondrial targeting as an option in cancer therapy.

2021 ◽  
Vol 28 ◽  
Author(s):  
Habibeh Mashayekhi-Sardoo ◽  
Adeleh Mashayekhi‐Sardoo ◽  
Basil D. Roufogalis ◽  
Tannaz Jamialahmadi ◽  
Amirhossein Sahebkar

: Curcumin, a yellow pigment in Asian spice, is a natural polyphenol component of Curcuma longa rhizome. Curcuminoid components include curcumin, demethoxycurcumin (DMC), and bisdemethoxycurcumin (BDMC). Previous studies established curcumin as a safe agent based on preclinical and clinical evaluations and curcuminoids have been approved by the US Food and Drug Administration (FDA) as “Generally Recognized as Safe” (GRAS). The present review collects and summarizes clinical and preclinical studies of curcumin interactions, with an emphasis on the effect of curcumin and curcumin analogs on the mRNA and protein levels of microsomal CYP450 enzymes (phase I metabolism) and their interactions with toxicants, drugs and drug probes. The literature search was conducted using keywords in various scientific databases, including Web of Science, Scopus, PubMed, and Google Scholar. Studies concerning the impact of curcumin and curcumin analogs on microsomal enzyme activity are reviewed and include oral, topical, and systemic treatment in humans and experimental animals, as well as studies from in vitro research. When taken together the data identified some inconsistent results between various studies. The findings showed significant inhibition of CYP450 enzymes by curcumin and its analogs. However such effects often differed when curcumin and curcumin analogs were coadministered with toxicant and other drugs and drug probes. We conclude from this review that herb-drug interactions should be considered when curcumin and curcumin analogs are consumed.


2021 ◽  
Author(s):  
Julia Bonnet ◽  
Lise Rigal ◽  
Odile Mondesert ◽  
Renaud Morin ◽  
Gaelle Corsaut ◽  
...  

Abstract Background Cancer cell aggregation is a key process involved in the formation of tumor cell clusters. It has recently been shown that clusters of circulating tumor cells (CTCs) have an increased metastatic potential compared to isolated circulating tumor cells. Several widely used chemotherapeutic agents that target the cytoskeleton microtubules and cause cell cycle arrest at mitosis have been reported to modulate CTC number or the size of CTC clusters. Results In this study, we investigated in vitro the impact of mitotic arrest on the ability of breast tumor cells to form clusters. By using live imaging and quantitative image analysis, we found that MCF-7 cancer cell aggregation is compromised upon incubation with paclitaxel or vinorelbine, two chemotherapeutic drugs that target microtubules. In line with these results, we observed that MCF-7 breast cancer cells experimentally synchronized and blocked in metaphase aggregated poorly and formed loose clusters. To monitor clustering at the single-cell scale, we next developed and validated an in vitro assay based on live video-microscopy and custom-designed micro-devices. The study of cluster formation from MCF-7 cells that express the fluorescent marker LifeAct-mCherry using this new assay allowed showing that substrate anchorage-independent clustering of MCF-7 cells was associated with the formation of actin-dependent highly dynamic cell protrusions. Metaphase-synchronized and blocked cells did not display such protrusions, and formed very loose clusters that failed to compact. Conclusions Altogether, our results suggest that mitotic arrest induced by microtubule-targeting anticancer drugs prevents cancer cell clustering and therefore, could reduce the metastatic potential of circulating tumor cells.


2019 ◽  
Vol 15 (11) ◽  
pp. 2151-2163 ◽  
Author(s):  
Lei Fang ◽  
Huaying Fan ◽  
Chunjing Guo ◽  
Linhan Cui ◽  
Peng Zhang ◽  
...  

Polymeric nanoparticles were widely used as delivery vehicles for targeted delivery of anticancer drugs, because of their targeting property and versatility. Mitochondria are one of the important organelles that regulate the apoptosis of cancer cells and can be considered as a pivotal target for cancer treatment. A pH-responsive charge-reversal and mitochondrial targeting nanoparticles, Vitamin B6-oligomeric hyaluronic acid-dithiodipropionic acid-berberine (B6-oHA-SS-Ber), were prepared in this study. Ber is a lipophilic cation that was conjugated with oHA through disulfide bonds to produce mitochondria-targeted conjugates (oHA-SS-Ber). B6 was conjugated to oHA to obtain B6-oHA-SS-Ber and the two types of Cur-loaded nanoparticles (Cur-NPs) were formulated by the dialysis method. Due to pKa of B6, the charge they carried in the tumor tissue acidic microenvironment can be transferred from negative charge to positive charge, further targeting mitochondria. In our study, we successfully synthesized B6-HA-SS-Ber and characterized the structure by 1H-NMR. According to the results of transmission electron microscopy (TEM), we found that the B6-oHA-SS-Ber/Cur micelles could self-assembled in water to form spherical nanoparticles, with a hydrodynamic diameter of 172.9±13 nm. Moreover, in vitro cytotoxicity, cellular uptake, lysosome escape and mitochondrial distribution researches revealed the better effect of B6-oHA-SS-Ber/Cur micelles in comparison to oHA-SS-Ber/Cur. In vivo anticancer activities indicated that the B6-oHA-SS-Ber/Cur micelles exhibited effective inhibition of tumor growth.


Cancers ◽  
2020 ◽  
Vol 12 (9) ◽  
pp. 2661
Author(s):  
Matti Ullah ◽  
Warda Aoudjeghout ◽  
Cynthia Pimpie ◽  
Marc Pocard ◽  
Massoud Mirshahi

Cancer is a result of “aggressive” division and uncontrolled proliferation of the abnormal cells that survive attack by immune cells. We investigated the expression of HLA-G and PD-L1 with the different stages of cancer cell division along with their role in the interaction of immune cells in vitro. Ovarian cancer (OVCAR-3) and chronic myeloid leukemia cell line (K-562) are used for this study. The correlation of protein expression with percentage of cells in each phase (G1, S and G2 phase) was evaluated through FACS. Cells were synchronized in G1, G2 and mitotic phase to evaluate gene (RT-qPCR) and protein expression (FACS). Real-time immune cell attack (RTICA) analysis with PBMCs (peripheral blood mono-nuclear cells) and cancer cells were performed. We found that cells expressing higher levels of HLA-G and PD-L1 are mainly in G2 phase and those expressing lower levels are mainly in G1 phase. Evidently, the higher expression of the two proteins was observed when synchronized in mitotic phase as compared to low expression when synchronized in G1 phase. RTICA analysis showed the presence of HLA-G delayed the lysis of the cells. In conclusion, the cancer cell can escape from immune cells in division stage that suggests the impact of mitosis index for cancer immunotherapy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3690-3690
Author(s):  
Takuya Tsubaki ◽  
Tetsuya Kadonosono ◽  
Tadashi Shiozawa ◽  
Takahiro Kuchimaru ◽  
Takashi Ushiki ◽  
...  

Abstract Introduction Solid tumors are infiltrated by a variety of myeloid-derived cells (MDCs), such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). Extensive studies have revealed that they play pivotal roles in tumor progression, such as immunosuppression, angiogenesis, and enhancing tumor cell invasion, and metastasis. It has been suggested, however, that undefined MDCs exist in tumors and play key roles in tumor progression. In order to develop a novel anti-tumor strategy, we have been searching undefined tumor-infiltrating MDCs that associate with tumor progression. Recently we have isolated adherent MDCs (AMCs), which strongly adhere to culture dishes, from subcutaneous tumors of Lewis lung carcinoma (LLC). AMCs contained CD45(+) CD11b(+) F4/80(-) undefined cell population (named F4/80(-)AMC) that exhibit protumoral functions. In this research, we characterized F4/80(-)AMC and explored its protumoral functions. Methods F4/80(-)AMC were subcutaneously transplanted into syngenic C57BL/6J mice with firefly luciferase-expressing LLC (LLC/luc) cells and bioluminescence (BL) signals corresponding to tumor burden were monitored every four days. The direct effect of F4/80(-)AMC on cancer cell growth was examined in vitro by culturing LLC/luc cells with (co-culture) or without (mono-culture) F4/80(-)AMC. In addition, conditioned medium (CM) collected from co-culture and mono-culture were analyzed by using mouse cytokine protein array. Receptors for markedly increased cytokines in co-culture CM were knocked down in LLC/luc cells to examine their involvement in LLC growth, and the candidate cytokines were further investigated for their direct effect on LLC growth by adding their recombinant proteins to the culture medium and monitoring the growth of LLC/luc cells. Furthermore, the possibility that F4/80(-)AMC could have a role to recruit other MDCs to tumor site was examined by in vitro chemotaxis assays using transwell chambers, and the impact of the neutralizing antibody against candidate cytokines on MDCs chemotaxis was evaluated. Results Investigation of surface makers on F4/80(-)AMC revealed that F4/80(-)AMC is distinct from any other already-known myeloid cells [Figure 1]. BL signal from tumors co-transplanted with F4/80(-)AMC was significantly increased compared to the signal from LLC/luc tumors, indicating that F4/80(-)AMC promotes tumor growth in subcutaneous tumor models [Figure 2]. Since F4/80(-) AMC was able to enhance proliferation of LLC cells in an in vitro co-culture without cell-cell contact [Figure 3], we hypothesized that F4/80(-)AMC may directly enhance tumor cell growth through cytokine release. Cytokine array showed that 6 cytokines (lipocalin-2, CXCL1, CXCL2, adiponectin, CCL2, and CCL5) were markedly increased in co-cultured CM compared to mono-cultured CM [Figure 4]. Knocking down (KD) of receptors for these cytokines indicate that only KD of CXCR2, a receptor for CXCL1/CXCL2, significantly abrogated F4/80(-)AMC-induced LLC growth [Figure 5]. CXCL1 and CXCL2 dose-dependently promoted LLC proliferation [Figure 6], demonstrating that F4/80(-)AMC directly enhance cancer cell proliferation via CXCL1 and CXCL2. Furthermore, in tumors co-transplanted with F4/80(-) AMC, monocytic MDSC (Mo-MDSC) and TAM were elevated, while activated CTLs were reduced [Figure 7]. It has been known that Mo-MDSC can suppress CTLs activities and that TAM is differentiated from Mo-MDSC in tumors. Therefore, above results may reflect F4/80(-)AMC-mediated recruitment of Mo-MDSC, which subsequently suppress CTLs and differentiate into TAM. Moreover, antibodies against CCL2 and CCL5 significantly suppressed the migration of Mo-MDSC toward CM of F4/80(-) AMC, suggesting that F4/80(-)AMC recruits Mo-MDSC via CCL2 and CCL5 secretion [Figure 8]. Taken together, these results strongly suggest that F4/80(-)AMC contributes to tumor progression by creating an immunosuppressive microenvironment. Conclusions Our study sheds light on the protumoral functions of novel MDCs: F4/80(−)AMC. Further characterization of F4/80(−)AMC and elucidation of its relationship with known MDCs are required to understand overall roles of MDCs in malignant progression. Our goal in this work is to identify the cell surface markers of F4/80(−)AMC and develop a novel treatment strategy for advanced cancers based on the knowledge of F4/80(−)AMC. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 16 (6) ◽  
pp. 853-866
Author(s):  
Olakunle Oladimeji ◽  
Jude Akinyelu ◽  
Moganavelli Singh

The mitochondria have recently become a novel target in the treatment of cancer. Targeted delivery by nanoparticles (NPs) has shown potential in enhancing existing therapeutic principles. With toxicity remaining a recurring issue, the green synthesis of inorganic NPs and modification with polymers may help to improve stability and biocompatibility. We synthesized epigallocatechin gallate (EGCG)-capped gold NPs (AuNPs), and functionalized with poly-D-lysine grafted polyethylene glycol (PDL-g-PEG), and the mitochondrial targeting triphenylphosphonium cation, and thereafter assessed their mitochondrial delivery capacity of paclitaxel in cancer cells in vitro. This PDL-g-PEG coated EGCG-AuNPs were further assessed for their laminin receptor avidity and mitochondrial localisation potential, upon functionalisation with the delocalised cation, triphenylphosphine. The laminin receptor dependent uptake and mitochondrial localisation of targeted T-Au(PDL-g-PEG) NPs were confirmed by ICP-OES and fluorescent microscopy. Their delivery of paclitaxel to the mitochondria of cancer cells elicited significant cytotoxicity especially in the human cervical carcinoma (HeLa) cell line, compared to the untargeted T-Au(PDL-g-PEG) and free drugs. Mechanistic studies implicated caspase dependent apoptosis as the mechanism of cell death. Our findings demonstrate the capacity of T-Au-[PDL-PEG] NPs to preferentially localize in the tumour mitochondria, and confirms the potential impact of subcellular targeting, especially to the mitochondria in cancer cells for an improvement in the therapeutic indices of these drugs.


2014 ◽  
Vol 10 (3) ◽  
pp. 473-479 ◽  
Author(s):  
Kai Jun Xiao ◽  
Ling Zhang ◽  
Xiao Hong Liu ◽  
Zhao Mei Wang ◽  
Shang Tian Yang

Abstract Cherokee rose fruit (CRF) is a Chinese traditional herb which has been used in medicine for hundreds of years. The anti-tumor activity of CRF polysaccharides (CRFPs) has not yet been evaluated. To study the in vitro anti-tumor effects of CRFP and its derivatives, native CRFP was isolated from CRF by hot water extraction, and its molecular weight analyzed with gel filtration chromatography was 227,000 Da. Native CRFP was sulfated with ClSO3H-DMF and carboxymethylated with monochloroacetic acid in alkaline aqueous medium. The resulting derivatives were isolated and labeled as SF-CRFP and CM-CRFP, respectively. The in vitro inhibition rates of CRFP and its derivatives for tumor cells SKVO (human ovarian cancer cell), HepG2 (human hepatoma cell), and LoVo (human colon cancer cell) were evaluated, the result showed that native CRFP exhibited no significant inhibition effect on the three tumor cells even at a concentration of 50 μg/ml, but sulfation and carboxymethylation substantially enhanced the anti-tumor activities of CRFP in a dose-dependent way. SF-CRFP at the dose of 50 μg/ml displayed a significant inhibitory effect on SKVO, HepG2, and LoVo, with the viability rates of 33.6%, 44.8%, and 43.2%, respectively. It has a dosage-dependence inhibition on tumor growth in this model, with IC50 for SKVO, HepG 2, and LoVo being 21 μg/ml, 36 μg/ml, and 49 μg/ml, respectively. CM-CRFP showed a specific inhibition on HepG2 with a viability rate of 12.2%, with an IC50 of 17 μg/ml, while it had hardly any anti-tumor effect on SKVO cells. Thus, chemical modifications of CRFPs by sulfation and carboxymethylation effectively improved their anti-tumor properties.


2020 ◽  
Author(s):  
Guiyang Wu ◽  
Chongshan Wu ◽  
Fubo Ye ◽  
Xiongwen Zhu ◽  
Zaiping Chen

Abstract Background Glucose metabolism transformation plays critical role in cancer cell malignancies maintenance. Aberrant cancer cell metabolism is considered to be the hallmark of cancer. S100A4 has been identified as an oncogene in a variety of cancers. However, its role in the cancer cell glucose reprogramming has been seldom reported. The aim of this study was to examine the role of S100A4 in aerobic glycolysis in colorectal cancer (CRC). Methods We investigated S100A4 expression in 224 cases of primary CRC and matched normal colonic tissue specimens, and explored the underlying mechanisms of altered S100A4 expression as well as the impact of this altered expression on CRC growth and glycolysis using in vitro and animal models of CRC. Results S100A4 was more highly expressed in CRC tissues than in the adjacent normal tissues (59.4% vs 17.4%, P <0.05). Higher S100A4 expression was associated with advanced node stage ( P =0.018) and larger tumor size ( P =0.035). A Cox proportional hazards model suggested that S100A4 expression was an independent prognostic factor for both OS (HR: 3.967, 95%CI: 1.919-8.200, P <0.001) and DFS (HR: 4.350, 95%CI: 2.264-8.358, P <0.001) in CRC after surgery. Experimentally, silencing S100A4 expression significantly decreased the growth and glycolysis rate of CRC both in vitro and in vivo . Mechanically, S100A4 could affect the hypoxia-inducible factor (HIF)-1α activity as demonstrated by the HIF-1α response element–luciferase activity in CRC cells. Conclusions These results disclose a novel role for S100A4 in reprogramming the metabolic process in CRC by affecting the HIF-1α activity and provide potential prognostic predictors for CRC.


2019 ◽  
Vol 17 (1) ◽  
pp. 685-693
Author(s):  
Chen Xi ◽  
Liu Yuanyuan ◽  
Zhao Dongshuang ◽  
Fan Ziwei ◽  
Cao Shuang ◽  
...  

AbstractIn this research, we investigated possible anti-inflammatory roles of Prunus tomentosa Thunb Total Flavones (PTTTF) in LPS-induced RAW264.7 cells. PTTTF (4μg/ml and 40μg/ml) was applied to RAW264.7 cells induced with 1μg/ml LPS to test the impact of these flavones on neutrophil phagocytosis in vitro. Levels of prostaglandin E2 (PGE2) and two pro-inflammatory interleukin cytokines (i.e. IL-6 and IL-1β) in the supernatant fraction were tested via Enzyme-linked immunosorbent assays (ELISA). Expression of cyclooxygenases COX-1 and COX-2 was detected via RT-PCR. Superoxide dismutase (SOD) content was determined with a spectrophotometric assay (Micromethod). The results revealed that PTTTF at doses higher than 4μg/ml reduces the content of IL-6, IL-1β and PGE2 (P < 0.05), and elevates the activity of SOD in LPS-induced RAW264.7 cells significantly (P < 0.05). PTTTF at 40μg/ml showed no significant effect on the expression of COX-1(P>0.05) but resulted in a significant inhibition of COX-2 in LPS-induced RAW264.7 cells (P<0.05). In summary, PTTTF had a substantial potential anti-inflammatory effect through the alteration of the synthesis of some cytokines and other mediators of the process of inflammation. Novelty statement - Prunus tomentosa Thunb Total Flavones (PTTTF) have known roles in the treatment of diabetes, but here we show that they are also potential anti-inflammatory agents. Our results show that PTTTF exhibited anti-inflammatory effects through altering the synthesis of some cytokines and other mediators of the inflammatory process.


Sign in / Sign up

Export Citation Format

Share Document