Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 5131-5144, 2024 07.
Article in English | MEDLINE | ID: mdl-38240783

ABSTRACT

This study explored the potential of poly-(lactic-co-glycolic) acid (PLGA) nanoparticles to enhance the effectiveness of anticancer treatments through combination therapy with phytol and α-bisabolol. The encapsulation efficiency of the nanoparticles was investigated, highlighting the role of ionic interactions between the drugs and the polymer. Characterization of PLGA-Phy+Bis nanoparticles was carried out using DLS with zeta potential and HR-TEM for size determination. Spectrophotometric measurements evaluated the encapsulation efficiency, loading efficiency, and in vitro drug release. FTIR analysis assessed the chemical interactions between PLGA and the drug actives, ensuring nanoparticle stability. GC-MS was employed to analyze the chemical composition of drug-loaded PLGA nanocarriers. Cytotoxicity was evaluated via the MTT assay, while Annexin V-FITC/PI staining and western blot analysis confirmed apoptotic cell death. Additionally, toxicity tests were performed on L-132 cells and in vivo zebrafish embryos. The study demonstrates high encapsulation efficiency of PLGA-Phy+Bis nanoparticles, which exhibit monodispersity and sizes of 189.3±5nm (DLS) and 268±54 nm (HR-TEM). Spectrophotometric analysis confirmed efficient drug encapsulation and release control. FTIR analysis revealed nanoparticle structural stability without chemical interactions. MTT assay results demonstrated the promising anticancer potential of all the three nanoparticle types (PLGA-Phy, PLGA-Bis, and PLGA-Phy+Bis) against lung cancer cells. Apoptosis was confirmed through Annexin V-FITC/PI staining and western blot analysis, which also revealed changes in Bax and Bcl-2 protein expression. Furthermore, the nanoparticles exhibited non-toxicity in L-132 cells and zebrafish embryo toxicity tests. PLGA-Phy+Bis nanoparticles exhibited efficient encapsulation, controlled release, and low toxicity. Apoptosis induction in A549 cells and non-toxicity in healthy cells highlight their clinical potential.


Subject(s)
Apoptosis , Drug Synergism , Lung Neoplasms , Monocyclic Sesquiterpenes , Nanoparticles , Phytol , Polylactic Acid-Polyglycolic Acid Copolymer , Zebrafish , Apoptosis/drug effects , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Humans , Nanoparticles/chemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Monocyclic Sesquiterpenes/pharmacology , Monocyclic Sesquiterpenes/administration & dosage , Phytol/administration & dosage , Phytol/pharmacology , Phytol/chemistry , Phytol/toxicity , Cell Line, Tumor , Drug Carriers/chemistry , A549 Cells , Drug Liberation , Sesquiterpenes/pharmacology , Sesquiterpenes/administration & dosage , Sesquiterpenes/chemistry , Sesquiterpenes/toxicity , Cell Survival/drug effects
2.
Eur J Pharmacol ; 949: 175699, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37011722

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer and the leading cause of gastrointestinal cancer death. 90% of people diagnosed with colorectal cancer are over the age of 50; nevertheless, the illness is more aggressive among those detected at a younger age. Chemotherapy-based treatment has several adverse effects on both normal and malignant cells. The primary signaling pathways implicated in the advancement of CRC include hedgehog (Hh), janus kinase and signal transducer and activator of transcription (JAK/STAT), Wingless-related integration site (Wnt)/ß-catenin, transforming growth factor-ß (TNF-ß), epidermal growth factor receptor (EGFR)/Mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K), nuclear factor kappa B (NF-κB), and Notch. Loss of heterozygosity in tumor suppressor genes like adenomatous polyposis coli, as well as mutation or deletion of genes like p53 and Kirsten rat sarcoma viral oncogene (KRAS), are all responsible for the occurrence of CRC. Novel therapeutic targets linked to these signal-transduction cascades have been identified as a consequence of advances in small interfering RNA (siRNA) treatments. This study focuses on many innovative siRNA therapies and methodologies for delivering siRNA therapeutics to the malignant site safely and effectively for the treatment of CRC. Treatment of CRC using siRNA-associated nanoparticles (NPs) may inhibit the activity of oncogenes and MDR-related genes by targeting a range of signaling mechanisms. This study summarizes several siRNAs targeting signaling molecules, as well as the therapeutic approaches that might be employed to treat CRC in the future.


Subject(s)
Colorectal Neoplasms , Phosphatidylinositol 3-Kinases , Humans , RNA, Small Interfering/genetics , Phosphatidylinositol 3-Kinases/metabolism , Hedgehog Proteins , Signal Transduction , ErbB Receptors/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics
3.
Int J Biol Macromol ; 202: 620-631, 2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35066022

ABSTRACT

Thymol is a plant-derived natural phenolic compound abundantly present in Thymus vulgaris species. In the present study, we developed a chitosan-based drug delivery system to deliver thymol to A549 cells. The physicochemical properties of thymol-loaded chitosan nanoparticles (thymol-NP) were characterized using polyphasic techniques viz., FTIR, XRD, DLS, and SEM. Thymol-NP exhibited a size of 282.5 nm and encapsulation efficiency of 74.08 ± 0.73%. The IC50 of thymol-NP against A549 cells was 99.57 µg/ml at 24 h, which was lower than that of the pure form. Clear apoptotic features such as cellular morphology, cell shrinkage, and augmentation of dead cells were observed in both the thymol and thymol-NP treated A549 cells. The percentage of apoptotic cells in the thymol-NP IC50 treated cells was >90% which was considerably higher than the group treated with thymol alone. In vivo toxicity study showed that the swiss albino mice treated up to a concentration of 1000 mg/kg of thymol-NP neither showed signs of toxicity nor death up to 14 days. Also, no significant influence was observed on behavior, body weight, organ weight, and organ histology. Overall, the data concluded that thymol-NP can be considered a safe and potent drug candidate against A549 cells.


Subject(s)
Chitosan , Nanoparticles , A549 Cells , Animals , Chitosan/chemistry , Drug Delivery Systems , Humans , Mice , Nanoparticles/chemistry , Thymol/chemistry , Thymol/pharmacology
4.
Pharmacol Rep ; 73(1): 240-254, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33095436

ABSTRACT

BACKGROUND: Thymol is a monoterpene phenol found in thyme species plants. The present study was carried out to investigate the effect of thymol and its molecular mechanism on non-small lung cancer (A549) cells. METHODS: The cytotoxic effect of thymol on A549 cells was assessed via MTT assay. ROS production, macromolecular damage, apoptosis were determined using DCF-DA, PI, AO/EtBr stains, respectively. ROS-dependent effect of thymol was confirmed using NAC. The expression of caspase-9, Bcl-2, Bax and cell cycle profile was analyzed via western blot and FACS, respectively. RESULTS: The antiproliferative effect of thymol on A549 cells was found to be both dose and time dependent with IC50 values of 112 µg/ml (745 µM) at 24 h. Thymol treatment favored apoptotic cell death and caused G0/G1 cell cycle arrest. It mediated cellular and nuclear morphological changes, phosphatidylserine translocation, and mitochondrial membrane depolarization. Additionally, upregulation of Bax, downregulation of Bcl-2, and apoptotic fragmented DNA were also observed. Thymol induced ROS by reducing the SOD level which was confirmed via in vitro and in silico analysis. Furthermore, the levels of lipid peroxides and protein carbonyl content were elevated in thymol-treated groups. Notably, N-acetyl cysteine pretreatment reversed the efficacy of thymol on A549 cells. Moreover, thymol-treated human PBMC cells did not show any significant cytotoxicity. CONCLUSION: Overall, our results confirmed that thymol can act as a safe and potent therapeutic agent to treat NSCLC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Thymol/pharmacology , A549 Cells , Acetylcysteine/pharmacology , Caspase 9/drug effects , Caspase 9/metabolism , Cell Cycle/drug effects , Computer Simulation , DNA Damage , Genes, bcl-2/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Thymol/antagonists & inhibitors , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
5.
Pharmacol Res ; 141: 73-84, 2019 03.
Article in English | MEDLINE | ID: mdl-30550953

ABSTRACT

JAK/STAT transduction pathway is a highly conserved pathway implicated in regulating cellular proliferation, differentiation, survival and apoptosis. Dysregulation of this pathway is involved in the onset of autoimmune, haematological, oncological, metabolic and neurological diseases. Over the last few years, the research of anti-neuroinflammatory agents has gained considerable attention. The ability to diminish the STAT-induced transcription of inflammatory genes is documented for both natural compounds (such as polyphenols) and chemical drugs. Among polyphenols, quercetin and curcumin directly inhibit STAT, while Berberis vulgaris L. and Sophora alopecuroides L extracts act indirectly. Also, the Food and Drug Administration has approved several JAK/STAT inhibitors (direct or indirect) for treating inflammatory diseases, indicating STAT can be considered as a therapeutic target for neuroinflammatory pathologies. Considering the encouraging data obtained so far, clinical trials are warranted to demonstrate the effectiveness and potential use in the clinical practice of STAT inhibitors to treat inflammation-associated neurodegenerative pathologies.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Nervous System Diseases/drug therapy , STAT Transcription Factors/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Humans , Inflammation/metabolism , Nervous System Diseases/metabolism , Polyphenols/pharmacology , Polyphenols/therapeutic use , STAT Transcription Factors/chemistry , STAT Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL