Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.467
Filter
1.
Oncol Res ; 32(7): 1197-1207, 2024.
Article in English | MEDLINE | ID: mdl-38948022

ABSTRACT

Breast cancer, a predominant global health issue, requires ongoing exploration of new therapeutic strategies. Palbociclib (PAL), a well-known cyclin-dependent kinase (CDK) inhibitor, plays a critical role in breast cancer treatment. While its efficacy is recognized, the interplay between PAL and cellular autophagy, particularly in the context of the RAF/MEK/ERK signaling pathway, remains insufficiently explored. This study investigates PAL's inhibitory effects on breast cancer using both in vitro (MCF7 and MDA-MB-468 cells) and in vivo (tumor-bearing nude mice) models. Aimed at elucidating the impact of PAL on autophagic processes and exploring the potential of combining it with trametinib (TRA), an MEK inhibitor, our research seeks to address the challenge of PAL-induced drug resistance. Our findings reveal that PAL significantly decreases the viability of MCF7 and MDA-MB-468 cells and reduces tumor size in mice while showing minimal cytotoxicity in MCF10A cells. However, PAL also induces protective autophagy, potentially leading to drug resistance via the RAF/MEK/ERK pathway activation. Introducing TRA effectively neutralized this autophagy, enhancing PAL's anti-tumor efficacy. A combination of PAL and TRA synergistically reduced cell viability and proliferation, and in vivo studies showed notable tumor size reduction. In conclusion, the PAL and TRA combination emerges as a promising strategy for overcoming PAL-induced resistance, offering a new horizon in breast cancer treatment.


Subject(s)
Autophagy , Breast Neoplasms , Piperazines , Pyridines , Pyridones , Pyrimidinones , Xenograft Model Antitumor Assays , Humans , Animals , Autophagy/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Female , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Mice , Piperazines/pharmacology , Piperazines/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Drug Synergism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mice, Nude , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cell Survival/drug effects , MCF-7 Cells
2.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000216

ABSTRACT

Astaxanthin (3,3'-dihydroxy-ß,ß-carotene-4,4'-dione; AXT) is a xanthophyll ß-carotenoid found in microalgae, seafood, fungi, complex plants, flamingos, and quail. It is well known that AXT plays a role as a drug with antioxidant and antitumor properties. Furthermore, several studies have reported that the reagent shows anti-inflammatory and neuroprotective effects. Recently, it was found that AXT acts as a peroxisome proliferator-activated receptor γ (PPARγ) modulator. To investigate the effect of AXT on MCF-7 cells (a human breast cancer cell line), the cells were treated with various concentrations of AXT. The treatment induced the decrease in cell number in a dose-dependent manner. Additionally, the Annexin V-positive cells were increased by the AXT treatment. These results indicated that apoptosis was induced in the tumor cells through the treatment of AXT. To elucidate the connection between apoptosis and p53, the levels of p53 and p21 proteins were assessed. Consequently, it was observed that the expression of p53 and p21 increased proportionally to the concentration of the AXT treatment. These findings suggest that the apoptosis of MCF-7 cells induced by AXT operates through a p53-dependent pathway, implying that AXT could potentially have a beneficial role in future breast cancer treatments. Thus, our results will provide a direction for future cancer challenges.


Subject(s)
Apoptosis , Signal Transduction , Tumor Suppressor Protein p53 , Xanthophylls , Humans , Tumor Suppressor Protein p53/metabolism , Xanthophylls/pharmacology , MCF-7 Cells , Apoptosis/drug effects , Signal Transduction/drug effects , Female , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism
3.
Methods Mol Biol ; 2842: 255-265, 2024.
Article in English | MEDLINE | ID: mdl-39012600

ABSTRACT

To fully exploit the potentials of reprogramming the epigenome through CRISPR/dCas9 systems for epigenetic editing, there is a growing need for improved transfection methods. With the utilization of constructs often with large sizes and the wide array of cell types used to read out the effect of epigenetic editing in different biological applications, it is evident that ongoing optimalization of transfection protocols tailored to each specific experimental setup is essential. Whether the goal is the production of viral particles using human embryonic kidney (HEK) cells or the direct examination of epigenomic modifications in the target cell type, continuous refinement of transfection methods is crucial. In the hereafter outlined protocol, we focus on optimization of transfection protocols by comparing different reagents and methods, creating a streamlined setup for transfection efficiency optimization in cultured mammalian cells. Our protocol provides a comprehensive overview of flow cytometry analysis following transfection not just to improve transfection efficiency but also to assess the expression level of the utilized construct. We showcase our transfection protocol optimization using HEK293T Lenti-X™ and breast cancer MCF-7 cell lines, using a single-guide RNA-containing plasmid. Specifically, we incorporate heat shock treatment for increased transfection efficiency of the MCF-7 cell line. Our detailed optimization protocol for efficient plasmid delivery and measurement of single-cell plasmid expression provides a comprehensive instruction for assessing both transient and sustained effects of epigenetic reprogramming.


Subject(s)
CRISPR-Cas Systems , Epigenesis, Genetic , Gene Editing , Plasmids , Single-Cell Analysis , Transfection , Humans , Plasmids/genetics , Gene Editing/methods , HEK293 Cells , Transfection/methods , Single-Cell Analysis/methods , Epigenomics/methods , Flow Cytometry
4.
Indian J Microbiol ; 64(2): 719-731, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39010984

ABSTRACT

Beta vulgaris var. crassa is undoubtedly a very important plant that is not used enough in the world. In this study, it was aimed to determine the cytotoxic activities of the components (essential oils, fatty acids, total phenol and flavonoid) found in the leaf parts of Beta vulgaris var. crassa against PC-3, MCF-7 and HeLa cancer cell lines. In addition, the effectiveness of these ingredients against bacteria and fungi that can cause serious health problems in humans was tested. In experiments, three tumor cell lines were exposed to various plant extract concentrations (31.25, 62.5, 125, 250, 500 and 1000 µg/mL) for 72 h. It was found that plant extracts showed high (SI: 2.14 > 2) cytotoxicity to PC-3 cells, moderate (SI: 1.62 < 2) to HeLa cells, and low (SI: 0.93 < 2) cytotoxicity to MCF-7 cells. Also, different plant extract concentrations were found to cause an inhibition rate of 16.3-22.3% in Staphylococcus aureus, 16.8-23.5% in Streptococcus pyogenes and 12-16.2% in Cutibacterium acnes. Similarly, inhibition rates were determined between 9.5-20.7% for Candida albicans, 3.5-7.7% for Candida auris, and 5.5-15.1% for Candida glabrata. The results showed that the plant extract exhibited a concentration-dependent cytotoxic and antimicrobial effect against both cancer cell lines and microbial pathogens. Supplementary Information: The online version contains supplementary material available at 10.1007/s12088-024-01269-8.

5.
Discov Oncol ; 15(1): 282, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008141

ABSTRACT

This study on Buddleja polystachya highlights its phytochemical composition, antimicrobial activity, and cytotoxic impacts. The study emphasizes the plant's potential to treat ocular diseases by identifying important compounds involved in the bioactivity through GC-MS analysis. This study explores the antimicrobial and cytotoxic potential of Buddleja polystachya (stem and leaves) extracts, with a focus on their application in treating bacterial ocular infections and their efficacy against MCF7, HT29, and HepG2 cancer cells. Through comprehensive GC-MS analysis, a diverse array of phytochemicals was identified within Buddleja polystachya stem and leaves extracts, including carbohydrates, phenolic derivatives, fatty acids, and steroidal components. The extracts were then evaluated for their biological activities, revealing significant antimicrobial properties against a range of bacterial strains implicated in ocular infections. The research findings demonstrate that stem extracts derived from Buddleja polystachya demonstrated high to moderate cytotoxic effects on cancer cell lines MCF7, HT29, and HepG2. Notably, these effects were characterized by varying IC50 values, which suggest distinct levels of sensitivity. In contrast, leaf extracts exhibited reduced cytotoxicity when tested against all these cell lines, although they did so with a significantly higher cytotoxicity aganist HepG2 cells. The results of this investigation highlight the potential therapeutic utilization of Buddleja polystachya extracts in the management of ocular infections and cancer. These results support the need for additional research to elucidate the underlying mechanisms of action of these extracts and explore their potential as drugs.

6.
Carbohydr Res ; 543: 109207, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39018698

ABSTRACT

Folic acid receptor-targeted drug delivery system is a promising candidate for tumor-targeted delivery because its elevated expression specifically on tumor cells enables the selective delivery of cytotoxic cargo to cancerous tissue, thereby minimizing toxic side effects and increasing the therapeutic index. Pyridine bisfolate-chitosan (PyBFA@CS NPs) and folate-chitosan nanocomposite (FA@CS NPs) were synthesized with suitable particle size (256.0 ± 15.0 and 161.0 ± 5.0 nm), high stability (ζ = -27.0 ± 0.1 and -30.0 ± 0.2 mV), respectively, and satisfactory biocompatibility to target cells expressing folate receptors and try to answer the question: Is the metal center always important for activity? Since almost all pharmaceuticals work by binding to specific proteins or DNA, the in vitro binding of human serum albumin (HSA) to PyBFA@CS NPs and FA@CS NPs has been investigated and compared with PyBFA. Strong affinity to HSA is shown by quenching and binding constants in the range of 105 and 104 M-1, respectively with PyBFA@CS NPs showing the strongest. The compounds-HSA kinetic stability, affinity, and association constants were investigated using a stopped-flow method. The findings showed that all formulations bind by a static quenching mechanism that consists of two reversible steps: rapid second-order binding and a more slowly first-order isomerization reaction. The overall coordination affinity of HSA to PyBFA@CS NPs (6.6 × 106 M-1), PyBFA (4.4 × 106 M-1), and FA@CS NPs (1.3 × 106 M-1) was measured and The relative reactivity is roughly (PyBFA@CS NPs)/(PyBFA)/(FA@CS NPs) = 5/3/1. Additionally, in vitro cytotoxicity revealed that, consistent with the binding constants and coordination affinity, active-targeting formulations greatly inhibited FR-positive MCF-7 cells in compared to FRs-negative A549 cells in the following trend: PyBFA@CS NPs > PyBFA > FA@CS NPs. Furthermore, in vitro drug release of PyBFA@CS NPs was found to be stable in PBS at pH 7.4, however, the in pH 5.4 and in pH 5.4 containing 10 mM glutathione (GSH) (mimicking the tumor microenvironment) reached 43 % and 73 %, respectively indicating that the PyBFA@CS NPs system is sensitive to GSH. Folate-modified nanoparticles, PyBFA@CS NPs, are a promising therapeutic for MCF-7 therapy because they not only showed a greater affinity for HSA, but also showed higher cleavage efficiency toward the minor groove of pBR322 DNA via the hydrolytic way, as well as effective antibacterial activity that avoids the usage of extra antibiotics.‬‬‬‬‬‬‬‬‬‬‬‬ ‬‬‬‬‬‬‬‬‬‬‬‬‬‬.

7.
Environ Res ; 259: 119445, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38942259

ABSTRACT

In present investigation, Carica papaya leaf extract has been employed as a bio-reductant agent in order to synthesize ecologically sustainable bio-coupled gold nanoparticles. The formation of gold nanoparticles was confirmed based on colour change of solution and its surface plasmon resonance peak measured using UV-Vis Spectrophotometer (UV-Vis). The Morphology and size of nanoparticles were determined using transmission electron microscope (SEM/TEM), and its crystalline structure by X-ray diffraction studies. Surface area was determined via BET isotherm analysis. The elemental composition of Au nanoparticles was developed using the technique of energy dispersive spectroscopy (EDS). Furthermore, FTIR analysis delineated the presence of functional groups present in the samples of the synthesized AuNPs. Thus, the efficiency of bio coupled Au nanoparticles in photo catalytically decomposing methylene blue was examined under the influence of visible light., the lethal MB colorant had been reduced to 95 % Within 90 min. And also 60% TOC removal was recorded after 5 min of degradation reaction, which increased to 99% after 90 min. Furthermore, cytotoxic experiments on Michigan Cancer Foundations-7 (MCF-7) cell lines showed that Au nanoparticles are effective anticancer agents with an IC50 of 87.2 g/mL on the top of the present work revealed the eco-safety and affordable production of Au nanoparticles from Carica papaya leaf extract, which displayed photocatalytic debasement of organic pollutants and cyto-toxicity effects was investigated.

8.
Molecules ; 29(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38930850

ABSTRACT

Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is a crucial tumor suppressor protein with frequent mutations and alterations. Although protein therapeutics are already integral to numerous medical fields, their potential remains nascent. This study aimed to investigate the impact of stable, unphosphorylated recombinant human full-length PTEN and its truncated variants, regarding their tumor suppression activity with multiwalled-carbon nanotubes (MW-CNTs) as vehicles for their delivery in breast cancer cells (T-47D, ZR-75-1, and MCF-7). The cloning, overexpression, and purification of PTEN variants were achieved from E. coli, followed by successful binding to CNTs. Cell incubation with protein-functionalized CNTs revealed that the full-length PTEN-CNTs significantly inhibited cancer cell growth and stimulated apoptosis in ZR-75-1 and MCF-7 cells, while truncated PTEN fragments on CNTs had a lesser effect. The N-terminal fragment, despite possessing the active site, did not have the same effect as the full length PTEN, emphasizing the necessity of interaction with the C2 domain in the C-terminal tail. Our findings highlight the efficacy of full-length PTEN in inhibiting cancer growth and inducing apoptosis through the alteration of the expression levels of key apoptotic markers. In addition, the utilization of carbon nanotubes as a potent PTEN protein delivery system provides valuable insights for future applications in in vivo models and clinical studies.


Subject(s)
Apoptosis , Breast Neoplasms , Cell Proliferation , Nanotubes, Carbon , PTEN Phosphohydrolase , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Nanotubes, Carbon/chemistry , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , MCF-7 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
9.
Antioxidants (Basel) ; 13(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38929147

ABSTRACT

Lycium barbarum, commonly recognized as goji berry or wolfberry, is highly appreciated not only for its organoleptic and nutritional properties but also as an important source of bioactive compounds such as polysaccharides, carotenoids, phenolics, and various other non-nutritive compounds. These constituents give it a multitude of health benefits, including antioxidant, anti-inflammatory, and anticancer properties. However, the precise biochemical mechanisms responsible for its anticancer effects remain unclear, and the comprehensive composition of goji berry extracts is often insufficiently explored. This study aimed to investigate the biochemical pathways modulated in breast cancer cells by an ethanolic extract of Lycium barbarum fruit (LBE). Following metabolomic profiling using UHPLC-HRMS/MS, we assessed the antitumoral properties of LBE on different breast cancer cell lines. This investigation revealed that LBE exhibited cytotoxic effects, inducing a pro-oxidant effect that triggered pyroptosis activation through endoplasmic reticulum (ER) stress and subsequent activation of the P-IRE1α/XBP1/NLRP3 axis in MCF-7 cells. In addition, LBE did not display cytotoxicity toward healthy human cells but demonstrated antioxidant properties by neutralizing ROS generated by doxorubicin. These findings underscore the potential of LBE as a highly promising natural extract in cancer therapy.

10.
Drug Dev Ind Pharm ; 50(6): 561-575, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38832870

ABSTRACT

INTRODUCTION: Breast cancer (BC) is the most common malignancy in women globally. Significant progress has been made in developing structural nanoparticles (NPs) and formulations for targeted smart drug delivery (SDD) of pharmaceuticals, improving the precision of tumor cell targeting in therapy. SIGNIFICANCE: Magnetic hyperthermia (MHT) treatment using magneto-liposomes (MLs) has emerged as a promising adjuvant cancer therapy. METHODS: CoFe2O4 magnetic NPs (MNPs) were conjugated with nanoliposomes to form MLs, and the anticancer drug quercetin (Que) was loaded into MLs, forming Que-MLs composites for antitumor approach. The aim was to prepare Que-MLs for DD systems (DDS) under an alternating magnetic field (AMF), termed chemotherapy/hyperthermia (chemo-HT) techniques. The encapsulation efficiency (EE), drug loading capacity (DL), and drug release (DR) of Que and Que-MLs were evaluated. RESULTS: The results confirmed successful Que-loading on the surface of MLs, with an average diameter of 38 nm and efficient encapsulation into MLs (69%). In vitro, experimental results on MCF-7 breast cells using MHT showed high cytotoxic effects of novel Que-MLs on MCF-7 cells. Various analyses, including cytotoxicity, apoptosis, cell migration, western blotting, fluorescence imaging, and cell membrane internalization, were conducted. The Acridine Orange-ethidium bromide double fluorescence test identified 35% early and 55% late apoptosis resulting from Que-MLs under the chemo-HT group. TEM results indicated MCF-7 cell membrane internalization and digestion of Que-MLs, suggesting the presence of early endosome-like vesicles on the cytoplasmic periphery. CONCLUSIONS: Que-MLs exhibited multi-modal chemo-HT effects, displaying high toxicity against MCF-7 BC cells and showing promise as a potent cytotoxic agent for BC chemotherapy.


Subject(s)
Apoptosis , Breast Neoplasms , DNA Damage , Hyperthermia, Induced , Liposomes , Quercetin , Humans , Quercetin/pharmacology , Quercetin/administration & dosage , Quercetin/chemistry , MCF-7 Cells , Apoptosis/drug effects , Hyperthermia, Induced/methods , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , DNA Damage/drug effects , Cobalt/chemistry , Cobalt/administration & dosage , Cobalt/pharmacology , Female , Ferric Compounds/chemistry , Drug Liberation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Drug Delivery Systems/methods , Magnetite Nanoparticles/chemistry , Cell Survival/drug effects , Magnetic Fields
11.
Drug Dev Res ; 85(4): e22224, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867474

ABSTRACT

The mammalian cytoplasmic protein SIRT2, a class III histone deacetylase family member, possesses NAD+-dependent lysine deacetylase/deacylase activity. Dysregulation of SIRT2 has been implicated in the pathogenesis of several diseases, including neurological and metabolic disorders and cancer; thus, SIRT2 emerges as a potential therapeutic target. Herein, we identified a series of diaryl acetamides (ST61-ST90) by the structural optimization of our hit STH2, followed by enhanced SIRT2 inhibitory potency and selectivity. Among them, ST72, ST85, and ST88 selectively inhibited SIRT2 with IC50 values of 9.97, 5.74, and 8.92 µM, respectively. Finally, the entire study was accompanied by in silico prediction of binding modes of docked compounds and the stability of SIRT2-ligand complexes. We hope our findings will provide substantial information for designing selective inhibitors of SIRT2.


Subject(s)
Acetamides , Sirtuin 2 , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/chemistry , Sirtuin 2/metabolism , Humans , Acetamides/chemistry , Acetamides/pharmacology , Molecular Docking Simulation , Structure-Activity Relationship , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry
12.
Int J Pharm ; 660: 124310, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38848796

ABSTRACT

Breast cancer has now replaced lung cancer as the most prevalent malignant tumor worldwide, posing a serious health risk to women. We have recently designed a promising option strategy for the treatment of breast cancer. In this work, cyclodextrin metal-organic frameworks with high drug-carrying properties were endo-crosslinked by 3,3'dithiodipropionyl chloride to form cubic phase gel nanoparticles, which were drug-loaded and then coated by MCF-7 cell membranes. After intravenous injection, this multifunctional nanomedicine achieved dramatically homologous targeting co-delivery of honokiol and indocyanine green to the breast tumor. Further, the disulfide bonds in the nanostructures achieved glutathione-responsive drug release, induced tumor cells to produce reactive oxygen species and promoted apoptosis, resulting in tumor necrosis, and at the same time, inhibited Ki67 protein expression, which enhanced photochemotherapy, and resulted in a 94.08 % in vivo tumor suppression rate in transplanted tumor-bearing mice. Thereby, this nanomimetic co-delivery system may have a place in breast cancer therapy due to its simple fabrication process, excellent biocompatibility, efficient targeted delivery of insoluble drugs, and enhanced photochemotherapy.


Subject(s)
Biphenyl Compounds , Breast Neoplasms , Drug Liberation , Glutathione , Indocyanine Green , Lignans , Metal-Organic Frameworks , Photochemotherapy , Indocyanine Green/administration & dosage , Indocyanine Green/chemistry , Animals , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , MCF-7 Cells , Photochemotherapy/methods , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/chemistry , Metal-Organic Frameworks/chemistry , Glutathione/metabolism , Lignans/administration & dosage , Lignans/chemistry , Lignans/pharmacology , Mice, Inbred BALB C , Cyclodextrins/chemistry , Mice , Apoptosis/drug effects , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Drug Delivery Systems/methods , Reactive Oxygen Species/metabolism , Mice, Nude , Drug Carriers/chemistry , Allyl Compounds , Phenols
13.
Article in English | MEDLINE | ID: mdl-38896333

ABSTRACT

PURPOSE: The insulin-like growth factor (IGF) system includes IGF-I, IGF-II insulin and their membrane receptors. IGF system also includes a family of proteins namely insulin-like growth factor-binding proteins (IGFBPs) composed for six major members (IGFBP-1 to IGFBP6), which capture, transport and prolonging half-life of IGFs. However, it has been described that IGFBPs can also have other functions. METHODS: IGFBP5 expression was inhibited by shRNAs, migration was analyzed by scratch-wound assays, invasion assays were performed by the Boyden chamber method, spheroids formation assays were performed on ultra-low attachment surfaces, expression and phosphorylation of proteins were analyzed by Western blot. RESULTS: IGFBP5 is a repressor of IGF-IR expression, but it is not a repressor of IR in MCF-7 breast cancer cells. In addition, IGFBP5 is a suppressor of migration and MMP-9 secretion induced by IGF-I and insulin, but it does not regulate invasion in MCF-7 cells. IGFBP5 also is a repressor of MCF-7 spheroids formation. However treatment with 340 nM rescues the inhibitory effect of IGFBP in the MCF-7 spheroids formation. CONCLUSION: IGFBP5 regulates IGF-IR expression, migration and MMP-9 secretion induced by IGF-I and/or insulin, and the spheroids formation in MCF-7 breast cancer cells.

14.
Transl Cancer Res ; 13(5): 2372-2386, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38881946

ABSTRACT

Background: Chidamide (CHI) is a subtype-selective histone deacetylase inhibitor (HDACI) developed in China and approved as a second-line treatment combined with the aromatase inhibitor for hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer. However, drug resistance is commonly occurred after a long period of medication. This study aimed to investigate the characterization of induced resistance to CHI and explore the potential cross-resistance to chemotherapeutic agents. Methods: CHI with gradually increasing concentrations was added to breast cancer MCF7 cells to establish a CHI-resistant MCF7 (MCF7-CHI-R) cell line. Cell counting kit-8 (CCK-8) assays were performed to detect half-maximal inhibitory concentration (IC50) of CHI. Colony formation was used to determine the proliferation inhibition rate. Western blot analysis was conducted to detect expressions of protein related with cell cycle, apoptosis, ferroptosis, and histone deacetylase (HDAC). Flow cytometry was used to analyze apoptosis and cell cycle. Results: The IC50 value of CHI of MCF7-CHI-R cells was increased in comparison with MCF7 cells. And CHI led to cell cycle arrest and ferroptosis, which were not exhibited in MCF7-CHI-R cells. Moreover, HDAC activity decreased in MCF7-CHI-R cells in comparison with MCF7 cells, and HDAC1 and HDAC10 might be involved in the resistance to CHI. In addition, MCF7-CHI-R cells were resistant to gemcitabine (GEM), doxorubicin (ADM), docetaxel (DXT), albumin-bound paclitaxel (nab-PTX) and paclitaxel (PTX). Conclusions: The MCF7-CHI-R was established and the anti-ferroptosis pathway activation was involved in the resistance of MCF-CHI-R cells. Also, MCF7-CHI-R cells were resistant to GEM, ADM, DXT, nab-PTX and PTX.

15.
Front Chem ; 12: 1384832, 2024.
Article in English | MEDLINE | ID: mdl-38887699

ABSTRACT

This study focused on developing new inhibitors for the MCF-7 cell line to contribute to our understanding of breast cancer biology and various experimental techniques. 3D QSAR modeling was used to design new tetrahydrobenzo[4, 5]thieno[2, 3-d]pyrimidine derivatives with good characteristics. Two robust 3D-QSAR models were developed, and their predictive capacities were confirmed through high correlations [CoMFA (Q2 = 0.62, R 2 = 0.90) and CoMSIA (Q2 = 0.71, R 2 = 0.88)] via external validations (R2 ext = 0.90 and R2 ext = 0.91, respectively). These successful evaluations confirm the potential of the models to provide reliable predictions. Six candidate inhibitors were discovered, and two new inhibitors were developed in silico using computational methods. The ADME-Tox properties and pharmacokinetic characteristics of the new derivatives were evaluated carefully. The interactions between the new tetrahydrobenzo[4, 5]thieno[2, 3-d]pyrimidine derivatives and the protein ERα (PDB code: 4XO6) were highlighted by molecular docking. Additionally, MM/GBSA calculations and molecular dynamics simulations provided interesting information on the binding stabilities between the complexes. The pharmaceutical characteristics, interactions with protein, and stabilities of the inhibitors were examined using various methods, including molecular docking and molecular dynamics simulations over 100 ns, binding free energy calculations, and ADME-Tox predictions, and compared with the FDA-approved drug capivasertib. The findings indicate that the inhibitors exhibit significant binding affinities, robust stabilities, and desirable pharmaceutical characteristics. These newly developed compounds, which act as inhibitors to mitigate breast cancer, therefore possess considerable potential as prospective drug candidates.

16.
J Adv Pharm Technol Res ; 15(2): 75-80, 2024.
Article in English | MEDLINE | ID: mdl-38903547

ABSTRACT

The invasive plant, Sphagneticola trilobata (L.) J. F. Pruski, has been known for its bioactivities and used to synthesize gold nanoparticles (AuNPs). Nonetheless, previous research has not directly compared the effectiveness of the plant parts in producing the AuNPs. The objective of this study was to compare the effectiveness of the flower and leaf of S. trilobata in synthesizing AuNPs. S. trilobata leaves and flowers were separately extracted using distilled water at 60°C for 30 min. The leaf and flower extracts were mixed with the HAuCl. 3H2O and heated to 60°C for 30 min to yield AuNPs-ALSt and AuNPs-AFSt, respectively. AuNPs were also prepared using trisodium citrate (Na3C6H5O7) as a control. The resultant AuNPs were characterized using an ultraviolet-visible spectrophotometer, particle size analyzer, and scanning electron microscope. Antioxidant activity was evaluated based on 1-diphenyl-2-picrylhydrazyl (DPPH) inhibition and anticancer activity- 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide assay against MCF-7 cells. The AuNPs-ALSt and AuNPs-AFSt were revealed to have better stability and smaller particle diameters. AuNPs-ALSt and AuNPs-AFSt had average particle diameters of 11.86 ± 3.37 and 34.86 ± 23.56 nm, respectively. Agglomeration was predominantly observed in AuNPs synthesized using the flower or leaf extract as stipulated to be affected by the insufficient capping agent and intense hydrolytic reaction. AuNPs-AFSt had higher DPPH antioxidant activity than AuNPs-ALSt with half-maximal inhibitory concentrations of IC50 123.44 and 168.83 ppm, respectively. Both AuNPs-ALSt and AuNPs-AFSt could inhibit 80% growth of the MCF-7; however, at lower concentrations, inhibitory effects were more pronounced in AuNPs-AFSt. Aqueous extracts of S. trilobata flowers and leaves could be used to synthesize AuNPs, whereas the former yielded AuNPs with higher biological activities.

17.
Sci Rep ; 14(1): 13744, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877067

ABSTRACT

Antitumor drugs used today have shown significant efficacy and are derived from natural products such as plants. Iso-mukaadial acetate (IMA) has previously been shown to possess anticancer properties by inducing apoptosis. The purpose of this study was to investigate the therapeutic effect of IMA in the breast cancer xenograft mice model. Female athymic nude mice were used and inoculated with breast cancer cells subcutaneously. Untreated group one served as a negative control and positive control group two (cisplatin) was administered intravenously. IMA was administered orally to group three (100 mg/kg) and group four (300 mg/kg). Blood was collected (70 µL) from the tail vein on day zero, day one and day three. Tumor regression was measured every second day and body mass was recorded each day. Estimation of serum parameters for renal indices was examined using a creatinine assay. Histopathological analysis was conducted to evaluate morphological changes of liver, kidney, and spleen tissues before and after compound administration under a fluorescence light microscope. Histopathological analysis of tumors was conducted before and after compound administration. Apoptotic analysis using the TUNEL system was conducted on liver, kidney, and spleen tissues. Tumor shrinkage and reduction in body mass were observed after treatment with IMA. Serum creatinine was slightly elevated after treatment with IMA at a dosage of 100 and 300 mg/kg. Histopathological results of the liver exhibited no changes before and after IMA while the kidney and spleen tissues showed changes in the cellular structure. IMA showed no cytotoxic effect on the tumor cells, and cell proliferation was observed. Apoptotic assay stain with TUNEL showed apoptotic cells in spleen tissue and kidney but no apoptotic cells were observed in liver tissue section treated with IMA. IMA showed clinical toxic signs that resulted in the suffering and death of the mice immediately after IMA administration. Histopathology of tumor cells showed that IMA did not inhibit cell proliferation and no cellular damage was observed. Therefore, based on the results obtained, we cannot make any definitive conclusion on the complete effect of IMA in vivo. IMA is toxic, poorly soluble, and not safe to use in animal studies. The objective of the study was not achieved, and the hypothesis was rejected.


Subject(s)
Apoptosis , Breast Neoplasms , Mice, Nude , Xenograft Model Antitumor Assays , Animals , Humans , Female , Mice , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Apoptosis/drug effects , MCF-7 Cells , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects
18.
Cell Biochem Biophys ; 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38850406

ABSTRACT

Cancer, defined by the continuous, uncontrollable proliferation of cells in the human body, is a disease with a rapidly increasing incidence and mortality rate. Scientists are looking for novel ways to cure and prevent this sneaky disease because of the toxicity of contemporary chemotherapy and the cancer cells' resilience to anticancer drugs. Determining the effect of herbal medicines, which do not have as harmful side effects as synthetic drugs, on cancer cell lines is an essential preliminary study in the production of effective drugs against cancer. In this study, the phenolic acid profile, antioxidant capacity, and cytotoxicity of the medicinal plant Mespilus germanica (MG) leaf extract were determined, and its effects on the expression of some apoptotic, necrotic, and autophagic pathway genes of MCF7 (Human breast cancer line) and A549 (Human lung cancer line) and healthy HDF (Human Dermal Fibroblasts) cells were investigated for the first time. The LCMS device detected many important phenolic compounds previously reported to act against cancer cells in Mespilus germanica leaf extract. DPPH and total phenolic content showed high antioxidant capacity. The cytotoxicity of MG was determined by the MTT method. The levels of mRNA transcription for Atg5, Atg3, Ripk1, Bcl2, Bax, Apaf1, Caspase-8, Caspase-7, Caspase-3, and Caspase-9, as well as the expression patterns of the DNA damage markers P53 and Parp-1 genes, were assessed. MG leaf extract did not cause significant toxicity against healthy HDF cells. However, it had a cytotoxic effect on A549 and MCF7 cancer cell lines, increasing the transcription levels of essential genes involved in cell death mechanisms. This research is the first to analyze the phenolic components and antioxidant capabilities of leaf extracts from Mespilus germanica. Additionally, it investigates the impact of these extracts on crucial genes involved in cell death pathways of A549 lung cancer, MCF7 breast cancer, and non-cancerous HDF (Human Dermal Fibroblasts) cells.

19.
Anal Chim Acta ; 1312: 342762, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38834277

ABSTRACT

Mucin1 (MUC1) is an extensively glycosylated transmembrane protein that is widely distributed and overexpressed on the surface of cancer cells, playing an important role in tumor occurrence and metastasis. Therefore, highly sensitive detection of MUC1 is of great significance for early diagnosis, treatment monitoring, and prognosis of cancer. Here, an ultra-sensitive photoelectrochemical (PEC) sensing platform was developed based on an aptamer amplification strategy for highly selective and sensitive detection of MUC1 overexpressed in serum and on cancer cell surfaces. The sensing platform utilized copper phthalocyanine to fabricate porous organic polymers (CuPc POPs), and was effectively integrated with g-C3N4/MXene to form a ternary heterojunction material (g-C3N4/MXene/CuPc POPs). This material effectively improved electron transfer capability, significantly enhanced light utilization, and greatly enhanced photoelectric conversion efficiency, resulting in a dramatic increase in photocurrent response. MUC1 aptamer 1 was immobilized on a chitosan-modified photoelectrode for the selective capture of MUC1 or MCF-7 cancer cells. When the target substance was present, MUC1 aptamer 2 labeled with methylene blue (MB) was specifically adsorbed on the electrode surface, leading to enhanced photocurrent. The concentration of MUC1 directly correlated with the number of MB molecules attracted to the electrode surface, establishing a linear relationship between photocurrent intensity and MUC1 concentration. The PEC biosensor exhibited excellent sensitivity for MUC1 detection with a wide detection range from 1 × 10-7 to 10 ng/mL and a detection limit of 8.1 ag/mL. The detection range for MCF-7 cells was from 2 × 101 to 2 × 106 cells/mL, with the capability for detecting single MCF-7 cells. The aptamer amplification strategy significantly enhanced PEC performance, and open up a promising platform to establish high selectivity, stability, and ultrasensitive analytical techniques.


Subject(s)
Aptamers, Nucleotide , Electrochemical Techniques , Mucin-1 , Polymers , Mucin-1/analysis , Humans , Aptamers, Nucleotide/chemistry , Electrochemical Techniques/methods , MCF-7 Cells , Porosity , Polymers/chemistry , Limit of Detection , Biosensing Techniques/methods , Indoles/chemistry , Photochemical Processes , Organometallic Compounds/chemistry
20.
J Inorg Biochem ; 258: 112617, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38805758

ABSTRACT

Four Pt(II) bis(pyrrole-imine) Schiff base chelates (1-4) were synthesised by previously reported methods, through a condensation reaction, and the novel crystal structure of 2,2'-{propane-1,3-diylbis[nitrilo(E)methylylidene]}bis(pyrrol-1-ido)platinum(II) (1) was obtained. Pt(II) complexes 1-4 exhibited phosphorescence, with increased luminescence in anaerobic solvents or when bound to human serum albumin (HSA). One of the complexes shows a 15.6-fold increase in quantum yield when bound to HSA and could be used to detect HSA concentrations as low as 5 nM. Pt(II) complexes 1-3 was investigated as potential theranostic agents in MCF-7 breast cancer cells, but only complex 3 exhibited cytotoxicity when irradiated with UV light (λ355nmExcitation). Interestingly, the cytotoxicity of complex 1 was unresponsive to UV light irradiation. This indicates that only complex 3 can be considered a potential photosensitising agent.


Subject(s)
Pyrroles , Humans , MCF-7 Cells , Pyrroles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Schiff Bases/chemistry , Imines/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/radiation effects , Coordination Complexes/chemical synthesis , Luminescent Agents/chemistry , Luminescent Agents/toxicity , Luminescent Agents/chemical synthesis , Breast Neoplasms/pathology , Breast Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...