scholarly journals Methotrexate Gold Nanocarriers: Loading and Release Study: Its Activity in Colon and Lung Cancer Cells

Molecules ◽  
2020 ◽  
Vol 25 (24) ◽  
pp. 6049
Author(s):  
Beatriz Álvarez-González ◽  
Marisa Rozalen ◽  
María Fernández-Perales ◽  
Miguel A. Álvarez ◽  
Manuel Sánchez-Polo

In the present study, the synthesis of gold nanoparticles (AuNPs) loaded with methotrexate (MTX) has been carried out in order to obtain controlled size and monodispersed nanocarriers of around 20 nm. The characterization study shows metallic AuNPs with MTX polydispersed on the surface. MTX is linked by the replacement of citrate by the MTX carboxyl group. The drug release profiles show faster MTX release when it is conjugated, which leads to the best control of plasma concentration. Moreover, the enhanced release observed at pH 5 could take advantage of the pH gradients that exist in tumor microenvironments to achieve high local drug concentrations. AuNP–MTX conjugates were tested by flow cytometry against lung (A-549) and colon (HTC-116) cancer cell lines. Results for A-549 showed a weaker dose–response effect than for colon cancer ones. This could be related to the presence of folate receptors in line HTC-116 in comparison to line A-549, supporting the specific uptake of folate-conjugated AuNP–MTX by folate receptor positive tumor cells. Conjugates exhibited considerably higher cytotoxic effects compared with the effects of equal doses of free MTX. Annexin V-PI tests sustained the cell death mechanism of apoptosis, which is normally disabled in cancer cells.

Author(s):  
Beatriz Álvarez-González ◽  
Marisa Rozalen ◽  
María Fernández-Perales ◽  
Miguel A. Álvarez ◽  
Manuel Sánchez-Polo

In the present study the synthesis of gold nanoparticles (AuNPs) loaded with methotrexate (MTX) has been carried out in order to obtain controlled size and monodispersed nanocarriers, around 20nm. Characterization study shows metallic AuNPs with MTX polydispersed on the surface. MTX is linked by a replacement of citrate by the MTX carboxyl group. The drug release profiles showed faster MTX release when it is conjugated, which leads to the best control of plasma concentration. Also, the enhanced release observed at pH 5 could take advantage of the pH gradients that exist in tumor microenvironments to achieve high local drug concentrations. AuNPs-MTX conjugates were tested by flow cytometry against lung (A-549) and colon (HTC-116) cancer cell lines. Results for A-549 showed a lighter dose-response effect than for colon cancer ones. This could be related to the presence of folate receptors in line HTC-116 on the contrary than line A-549, supporting the specific uptake of folate-conjugated AuNPs-MTX by folate receptor positive tumor cells. Conjugates exhibited considerably higher cytotoxic effects compared with the effects of equal doses of free MTX. Anexin V-PI test sustain as cell death mechanism apoptosis, which is normally disabled in cancer cells.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3981-3981 ◽  
Author(s):  
Mariola Sadowska ◽  
Nidal Muvarak ◽  
Rena G. Lapidus ◽  
Edward A. Sausville ◽  
Rajat Bannerji ◽  
...  

Abstract Abstract 3981 Dinaciclib (SCH 727965) is a selective and potent inhibitor of CDK 1, 2, 5 and 9 (IC50 < 5 nM) that has demonstrated in vitro and in vivo anti-tumor activity against a variety of tumor cell lines and human tumor xenograft models. The concentration of dinaciclib required to achieve these effects (< 100 nM) is achieved in clinical trials, and dinaciclib was found to have a more favorable therapeutic index, in preclinical murine models, than other CDK inhibitors. We have studied the effect of dinaciclib on human acute myelogenous (AML) and acute lymphoblastic (ALL) leukemia cell lines, including HL-60, K562 and Molt-4, and primary leukemia cells. Dose response curves (0.0004-10 μM) were generated for different exposure times (2, 24 and 72 h), and data from cell proliferation assay (WST-1) were used to calculate the IC50 values. Short 2 h exposure to dinaciclib followed by 24 h culture without drug resulted in different responses between the cell lines (IC50 values of 0.13 μM, 2.17 μM and ND; and viability at 10 μM 62%, 76% and 95%, for HL-60, Molt-4, and K562, respectively). With longer exposure times (24 and 72 h), the IC50 was similar between the cell lines (IC50 24 h values of 0.017, 0.015, and 0.019 μM for HL-60, Molt-4, and K562, respectively). However, even in the presence of the highest drug concentration tested (10 μM), approximately 5–25% of cells remained metabolically active after 24 h culture, and in a colony forming assay were able to proliferate and form colonies after removal of the drug. Longer 72 h exposure to dinaciclib (0.2-10 μM) completely inhibited cell proliferation in all cell lines and prevented colony formation. Next, we examined the effect of dinaciclib (2-200 nM) on cell cycle in HL-60 and K562 cells (2, 6, 9, 24 h). While lower drug concentrations and shorter exposures resulted in a minor increase in the proportion of cells in the G2 phase, a considerable increase of cells in the sub-G1 phase was observed with prolonged exposures and higher drug concentrations, most prominently in HL-60 cells (4h 200 nM 38%; 6h 20 nM 53% or 200 nM 71%, and 24 h 20 nM 84%), which is consistent with cell viability assay data. These findings were also confirmed by Annexin V/PI staining. To characterize the molecular mechanisms behind the induction of cell cycle arrest and apoptosis by dinaciclib, we measured the changes in protein expression of Mcl-1, phosphorylation of retinoblastoma (Rb) protein, and cleavage of PARP by Western blotting. Dinaciclib treatment in a dose- and time-dependent manner (6 and 24 h; 10–500 nM) significantly decreased the expression of anti-apoptotic protein Mcl-1, Rb phosphorylation at Ser 811/817, and induced cleavage of the PARP protein in the three cell lines tested. For HL-60 cells, even 2 h exposure to dinaciclib was able to induce these effects when cells were examined 4 h after treatment; however, both Mcl-1 and p-Rb returned to baseline 24 h later, suggesting that the cells were able to recover. Using HL-60 cells, we were also able to demonstrate that a decrease in Mcl-1 correlates with the decrease in phosphorylation of the carboxy-terminal domain of RNA polymerase II, suggesting that dinaciclib successfully inhibits CDK9 which may lead to transcriptional down-regulation of Mcl-1. Dinaciclib treatment also down-regulated the expression of XIAP, Bcl-xl, and phosphorylation of Bad at Ser 112 (the pro-survival form of Bad), while Bak and Bax levels remained unaffected. The cleavage of PARP correlated with the activation of the caspase-3 and -9, suggesting the involvement of the intrinsic pathway of apoptosis. We confirmed our findings in primary leukemia cells. Dinaciclib was able to induce growth inhibition in all 7 primary AML samples (IC50 for 24 h exposure ranging from 0.008 to 0.017 μM) and apoptosis (Annexin V/PI staining). Treatment with dinaciclib also resulted in down-regulation of Mcl-1, cleavage of PARP, and dephosphorylation of Rb in all primary leukemia cells examined. In summary, dinaciclib potently inhibits the growth and induces apoptosis of human leukemia cells in vitro. Prolonged exposure times may be required for its maximum efficacy, and given its short half-life in humans (1.5 to 3.3 hours), this should be considered when designing the clinical studies for patients with acute leukemias. Disclosures: Sadowska: Merck & Co: Research Funding. Muvarak:Merck & Co: Research Funding. Lapidus:Merck & Co: Equity Ownership, Research Funding. Bannerji:Merck & Co: Employment, Equity Ownership. Gojo:Merck & Co.: Research Funding.


2019 ◽  
Vol 7 (39) ◽  
pp. 5983-5991 ◽  
Author(s):  
Zihou Li ◽  
Xuehua Ma ◽  
Yuanzhi Xia ◽  
Kun Qian ◽  
Ozioma Udochukwu Akakuru ◽  
...  

A T2-weighted MRI contrast agent (SPION-AN-FA@mPEG) can precisely target cancer cells with folate receptor α (FRα) diminishing non-specific uptake by normal healthy cells.


2018 ◽  
Vol 18 (7) ◽  
pp. 1054-1063 ◽  
Author(s):  
Ning Ding ◽  
Hong Zhang ◽  
Shan Su ◽  
Yumei Ding ◽  
Xiaohui Yu ◽  
...  

Background: Endometrial cancer is a common cause of death in gynecological malignancies. Cisplatin is a clinically chemotherapeutic agent. However, drug-resistance is the primary cause of treatment failure. Objective: Emodin is commonly used clinically to increase the sensitivity of chemotherapeutic agents, yet whether Emodin promotes the role of Cisplatin in the treatment of endometrial cancer has not been studied. Method: CCK-8 kit was utilized to determine the growth of two endometrial cancer cell lines, Ishikawa and HEC-IB. The apoptosis level of Ishikawa and HEC-IB cells was detected by Annexin V / propidium iodide double-staining assay. ROS level was detected by DCFH-DA and NADPH oxidase expression. Expressions of drug-resistant genes were examined by real-time PCR and Western blotting. Results: Emodin combined with Cisplatin reduced cell growth and increased the apoptosis of endometrial cancer cells. Co-treatment of Emodin and Cisplatin increased chemosensitivity by inhibiting the expression of drugresistant genes through reducing the ROS levels in endometrial cancer cells. In an endometrial cancer xenograft murine model, the tumor size was reduced and animal survival time was increased by co-treatment of Emodin and Cisplatin. Conclusion: This study demonstrates that Emodin enhances the chemosensitivity of Cisplatin on endometrial cancer by inhibiting ROS-mediated expression of drug-resistance genes.


2019 ◽  
Vol 19 (15) ◽  
pp. 1835-1845
Author(s):  
Ali Hassanzadeh ◽  
Adel Naimi ◽  
Majid F. Hagh ◽  
Raedeh Saraei ◽  
Faroogh Marofi ◽  
...  

Introduction: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL or Apo2L) is a member of the tumor necrosis factor (TNF) superfamily, which stimulates apoptosis in a wide range of cancer cells via binding to death receptors 4 and 5 (DR4/5). Nevertheless, TRAIL has noticeable anti-cancer abilities; some cancer cells acquire resistance to TRAIL, and consequently its potential for inducing apoptosis in target cells is strongly diminished. Acute lymphoblastic leukemia MOLT-4 cell line is one of the most resistant cells to TRAIL that developed resistance to TRAIL via different pathways. We used TRAIL plus kaempferol to eliminate resistance of the MOLT-4 cells to TRAIL. Material and Methods: First, IC50 for kaempferol (95 µM) was determined by using the MTT assay. Second, the viability of the MOLT-4 cells was assayed by FACS after Annexin V/PI staining, following treatment with TRAIL (50 and 100 nM) and kaempferol (95 µM) alone and together. Finally, the expression levels of the candidate genes involved in resistance to TRAIL were assayed by real-time PCR technique. Results: Kaempferol plus TRAIL induced apoptosis robustly in MOLT-4 cells at 12, 24 and 48 hours after treatment. Additionally, we found that kaempferol could inhibit expression of the c-FLIP, X-IAP, cIAP1/2, FGF-8 and VEGF-beta, and conversely augment expression of the DR4/5 in MOLT-4 cells. Conclusion: We suggest that co-treatment of MOLT-4 cells with TRAIL plus kaempferol is a practical and attractive approach to eliminate cancers’ resistance to TRAIL via inhibition of the intracellular anti-apoptotic proteins, upregulation of DR4/5 and also by suppression of the VEGF-beta (VEGFB) and FGF-8 expressions.


2019 ◽  
Vol 19 (6) ◽  
pp. 826-837 ◽  
Author(s):  
Pratibha Pandey ◽  
Preeti Bajpai ◽  
Mohammad H. Siddiqui ◽  
Uzma Sayyed ◽  
Rohit Tiwari ◽  
...  

Background:Plant sterols have proven a potent anti-proliferative and apoptosis inducing agent against several carcinomas including breast and prostate cancers. Jab1 has been reported to be involved in the progression of numerous carcinomas. However, antiproliferative effects of sterols against Jab1 in gall bladder cancer have not been explored yet.Objective:In the current study, we elucidated the mechanism of action of stigmasterol regarding apoptosis induction mediated via downregulation of Jab1 protein in human gall bladder cancer cells.Methods:In our study, we performed MTT and Trypan blue assay to assess the effect of stigmasterol on cell proliferation. In addition, RT-PCR and western blotting were performed to identify the effect of stigmasterol on Jab1 and p27 expression in human gall bladder cancer cells. We further performed cell cycle, Caspase-3, Hoechst and FITC-Annexin V analysis, to confirm the apoptosis induction in stigmasterol treated human gall bladder cancer cells.Results:Our results clearly indicated that stigmasterol has up-regulated the p27 expression and down-regulated Jab1 gene. These modulations of genes might occur via mitochondrial apoptosis signaling pathway. Caspase-3 gets activated with the apoptotic induction. Increase in apoptotic cells and DNA were confirmed through annexin V staining, Hoechst staining, and cell cycle analysis.Conclusion:Thus, these results strongly suggest that stigmasterol has the potential to be considered as an anticancerous therapeutic agent against Jab1 in gall bladder cancer.


2021 ◽  
pp. 096032712110214
Author(s):  
JY Lee ◽  
HM Lim ◽  
CM Lee ◽  
S-H Park ◽  
MJ Nam

Indole-3-carbinol (I3C) is a phytochemical that exhibits growth-inhibitory activity against various cancer cells. However, there are limited studies on the effects of I3C on colon cancer cells. In this study, the growth-inhibitory activity of I3C against the human colorectal carcinoma cell line (LoVo) was examined. The results of the 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide, colony formation, and cell counting assays revealed that I3C suppressed the proliferation of LoVo cells. Microscopy and wound-healing analyses revealed that I3C affected the morphology and inhibited the migration of LoVo cells, respectively. I3C induced apoptosis and DNA fragmentation as evidenced by the results of fluorescein isothiocyanate-conjugated annexin V staining and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling assay, respectively. Additionally, I3C arrested the cell cycle at the G0/G1 phase and enhanced the reactive oxygen species levels. Western blotting analysis revealed that treatment with I3C resulted in the activation of apoptotic proteins, such as poly(ADP-ribose) polymerase, caspase-3, caspase-7, caspase-9, Bax, Bim, and p53 in LoVo cells. These results indicate that I3C induces apoptosis in LoVo cells by upregulating p53, leading to the activation of Bax and caspases. Taken together, I3C exerts cytotoxic effects on LoVo cells by activating apoptosis.


2021 ◽  
Vol 7 (3) ◽  
pp. 188
Author(s):  
Ji-In Noh ◽  
Seul-Ki Mun ◽  
Eui Hyeon Lim ◽  
Hangun Kim ◽  
Dong-Jo Chang ◽  
...  

Physconia hokkaidensis methanol extract (PHE) was studied to identify anticancer effects and reveal its mechanism of action by an analysis of cytotoxicity, cell cycles, and apoptosis biomarkers. PHE showed strong cytotoxicity in various cancer cells, including HL-60, HeLa, A549, Hep G2, AGS, MDA-MB-231, and MCF-7. Of these cell lines, the growth of MDA-MB-231 was concentration-dependently suppressed by PHE, but MCF-7 was not affected. MDA-MB-231 cells, triple-negative breast cancer (TNBC) cells, do not express estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2), whereas MCF-7 cells are ER-positive, PR-positive, and HER-2-negative breast cancer cells. The number of cells in sub-G1 phase was increased after 24 h of treatment, and annexin V/PI staining showed that the population size of apoptotic cells was increased by prolonged exposure to PHE. Moreover, PHE treatment downregulated the transcriptional levels of Bcl-2, AMPK, and p-Akt, whereas it significantly upregulated the levels of cleaved caspase-3, cleaved caspase-9, and cleaved-PARP. In conclusion, it was confirmed that the PHE exhibited selective cytotoxicity toward MDA-MB-231, not toward MCF-7, and its cytotoxic activity is based on induction of apoptosis.


2020 ◽  
Vol 9 (1) ◽  
pp. 192 ◽  
Author(s):  
Alexandre Quilbe ◽  
Olivier Moralès ◽  
Martha Baydoun ◽  
Abhishek Kumar ◽  
Rami Mustapha ◽  
...  

To date, pancreatic adenocarcinoma (ADKP) is a devastating disease for which the incidence rate is close to the mortality rate. The survival rate has evolved only 2–5% in 45 years, highlighting the failure of current therapies. Otherwise, the use of photodynamic therapy (PDT), based on the use of an adapted photosensitizer (PS) has already proved its worth and has prompted a growing interest in the field of oncology. We have developed a new photosensitizer (PS-FOL/PS2), protected by a recently published patent (WO2019 016397-A1, 24 January 2019). This photosensitizer is associated with an addressing molecule (folic acid) targeting the folate receptor 1 (FOLR1) with a high affinity. Folate binds to FOLR1, in a specific way, expressed in 100% of ADKP or over-expressed in 30% of cases. The first objective of this study is to evaluate the effectiveness of this PS2-PDT in four ADKP cell lines: Capan-1, Capan-2, MiapaCa-2, and Panc-1. For this purpose, we first evaluated the gene and protein expression of FOLR1 on four ADKP cell lines. Subsequently, we evaluated PS2’s efficacy in our cell lines and we assessed the impact of PDT on the secretome of cancer cells and its impact on the immune system. Finally, we evaluate the PDT efficacy on a humanized SCID mouse model of pancreatic cancer. In a very interesting way, we observed a significant increase in the proliferation of activated-human PBMC when cultured with conditioned media of ADKP cancer cells subjected to PDT. Furthermore, to evaluate in vivo the impact of this new PS, we analyzed the tumor growth in a humanized SCID mice model of pancreatic cancer. Four conditions were tested: Untreated, mice (nontreated), mice with PS (PS2), mice subjected to illumination (Light only), and mice subjected to illumination in the presence of PS (PDT). We noticed that the mice subjected to PDT presented a strong decrease in the growth of the tumor over time after illumination. Our investigations have not only suggested that PS2-PDT is an effective therapy in the treatment of PDAC but also that it activates the immune system and could be considered as a real adjuvant for anti-cancer vaccination. Thus, this new study provides new treatment options for patients in a therapeutic impasse and will provide a new arsenal in the fight against PDAC.


Sign in / Sign up

Export Citation Format

Share Document