فهرست مطالب
International Journal of Molecular and Cellular Medicine
Volume:13 Issue: 51, Summer 2024
- تاریخ انتشار: 1403/10/23
- تعداد عناوین: 8
-
-
Pages 220-233
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with a poor response to the limited treatment options currently available. Hence, there is a need to identify new agents that could enhance the efficacy of existing treatments. This study investigated a combination therapy using gemcitabine (GEM) and SCH772984, an extracellular signal-regulated kinase (ERK) inhibitor, in both free form and nanoparticle-encapsulated form for PDAC treatment. Cell viability and Matrigel growth assays were used to determine the anti-proliferative and cytotoxic effects of GEM and SCH772984 on PDAC cells. Additionally, western blotting was used to determine the degree to which SCH772984 engaged ERK in PDAC cells. Lastly, immunohistochemistry and hematoxylin and eosin (H&E) staining were used to determine how GEM and SCH772984 affected expression of Ki-67 cell proliferation marker in PDX (patient derived xenograft) PDAC tissues. PDAC cell lines (MIA PaCa-2 and PANC-1) treated with the combination of free GEM and SCH772984 showed reduction in cell viability compared to cells treated with free GEM or SCH772984 administered as a single agent. Encapsulated forms of GEM and SCH772984 caused a greater reduction in cell viability than the free forms. Interestingly, co-administration of GEM and SCH772984 in separate nanoparticle (NP) systems exhibited the highest reduction in cell viability. Western blotting analysis confirmed ERK signaling was inhibited by both free and encapsulated SCH772984. Importantly, GEM did not interfere with the inhibitory effect of SCH772984 on phosphorylated ERK (pERK). Collectively, our studies suggest that combination therapy with GEM and SCH772984 effectively reduced PDAC cell viability and growth, and co-administration of NP encapsulated GEM and SCH772984 in separate NP systems is an effective treatment strategy for PDAC.
Keywords: ERK Inhibitor, Gemcitabine, Nanoparticle, PDAC -
Pages 234-247
Transforming growth factor beta (TGF-β) initiates epithelial-mesenchymal transition (EMT) in tubular and glomerular epithelial cells, resulting in excessive production and deposition of extracellular matrix through its interaction with TGF-β receptors, which play a crucial role in TGF-β signaling involving two receptor types, namely TGF-β type I (TβRI) and type II (TβRII). EMT contributes to the pathogenesis of interstitial renal fibrosis, a marker of end-stage kidney disease. This study aimed to identify the bioactive compounds in the active fraction of P. angulata and evaluate their ability to inhibit the TGF-β activity and their potential as drug candidates. The active components in the active fraction of P. angulata were analyzed using gas chromatography-mass spectrometry (GC-MS). The bioactive compound structures were obtained from the PubChem database, while the protein targets, TβRI and TβRII, were retrieved from the Protein Data Bank (PDB). The molecular docking analyses were performed using PyRx 0.8 and Discovery Studio. SwissADME was used to evaluate ligand properties and druglikeness. Three dominant active compounds were identified, namely palmitic acid, campesterol, and stigmasterol. In silico studies demonstrated strong energy bonds existed between TβRI and palmitic acid, campesterol, stigmasterol, and SB431542 with binding energy values of -5.7, -10, -9.4, and -10.9 kcal/mol, respectively. Similarly, they strongly bound to TβRII with binding energy values of -5.2, -7.1, -7.5, and -6.1 kcal/mol, respectively. All compounds meet Lipinski’s criteria for druglikeness. Among the identified active compounds, campesterol exhibited the highest affinity for TβRI, while stigmasterol exhibited a strong affinity for TβRII. These findings suggested that the three compounds have potential as drug candidates.
Keywords: Diabetic Nephropathy, Transforming Growth Factor Beta, TGF-Β Inhibitor, Physalis Angulata -
Pages 248-258
Chronic lymphocytic leukemia (CLL) is the most prevalent hematological cancer, with various medical interventions. In the recent decade, cold physical plasma has become an interesting agent for future cancer therapy. The goal of this study was to see whether cold physical plasma or cold physical plasma-treated liquid (PTL) affected integrin beta 3 (ITGB3) expression, which is hypothesized to mediate an interaction between cancer stem cells and the bone marrow microenvironment, in CLL patients' blood cells. The metabolic activity, cell death pattern, lipid oxidation and ITGB3 gene expression of these treatments was evaluated. Both direct cold physical plasma and PTL exposure enhanced lipid peroxidation in cells of CLL patients, but to a lesser extent in healthy participants. Furthermore, following 48h of cold physical plasma or PTL exposure, the metabolic activity of leukocytes was preferentially reduced in CLL patient leukocytes. In addition, cold physical plasma and PTL treatment elevated ITGB3 mRNA expression in CLL patients' leukocytes compared to untreated and healthy controls. Collectively, our study suggests selective effects of direct cold physical plasma and PTL exposure on blood leukocytes from leukemia patients, but further and more detailed studies are needed to provide additional rationales for such treatment options as future therapy.
Keywords: CAP, Cold Atmospheric Pressure Plasma, Chronic Lymphocytic Leukemia, Gas Plasma Technology, NK Cells, Reactive Oxygen Species, ROS -
Pages 259-271
Overexpression of (myeloid leukemia cell differentiation protein 1) Mcl-1 is associated with the reduction of ABT-737 toxicity and secondary resistance. In this study, the effect of formononetin (biochanin B) on Mcl-1 expression, cell growth, apoptosis and ABT-737 sensitivity of the acute lymphoblastic leukemia (ALL) cells was investigated. In this experimental study, the cell proliferation and MTT assays were used to investigate the effect of formononetin on cell growth and survival. qRT-PCR was performed for the measurement of gene expression. Hoechst 33342 staining and caspase-3 activity assay were used for the determination of apoptosis. Our data showed that formononetin and ABT-737 both led to a significant reduction in the IC50 value and synergistically reduced the cell growth and survival relative to single treatment. Overexpression of Mcl-1 was found after the treatment with ABT-737. Formononetin decreased the expression of B-cell lymphoma 2 (Bcl-2) and Mcl-1 and increased the Bcl-2-associated protein x (Bax) and P21 expression. Moreover, formononetin enhanced the apoptotic effect of ABT-737 in ALL cells. In summary, formononetin showed anti-carcinogenic activities in human ALL cells via suppression of cell growth and survival. Formononetin enhanced the apoptotic effect of ABT-737, with contribution by inhibition of the Mcl-1 expression.
Keywords: ABT-737, ALL, Apoptosis, Formononetin, Mcl-1 -
Pages 272-285
Breast cancer, characterized by genetic diversity and molecular subtypes, presents significant treatment challenges, especially in human epidermal growth factor receptor type 2 (HER2)-positive cases, which are associated with poor prognosis. Metformin, widely known for its antidiabetic effects, has emerged as a promising candidate for cancer therapy. This study investigates the effect of metformin on miR-125a promoter methylation and its subsequent impact on the HER2 signaling pathway in HER2-positive breast cancer cells (SK-BR3). SK-BR3 cells were cultured and treated with various concentrations of metformin to assess its effects on cell viability, DNA methylation, HER2, and DNA Methyltransferase 1 (DNMT1) expression. Molecular analyses focus on the miR-125a signaling pathway modulation, DNA methylation, mRNA expression of DNMT1, and protein level of HER2. Research showed a dose-dependent reduction in cell viability, with IC50 values from 65 mM at 48 hours to 35 mM at 72 hours. Metformin treatment led to demethylation of the miR-125a promoter, which increased miR-125a expression and subsequently reduced HER2 levels. This suggests that metformin exerts its anticancer effects partly by regulation of the miR-125a-HER2 axis. Additionally, metformin inhibited vimentin expression, indicating its potential to interfere with epithelial-mesenchymal transition (EMT) processes. Metformin may serve as a targeted therapeutic agent in HER2-positive breast cancer by modulating the miR-125a-HER2 axis and influencing on the epigenetic and EMT regulation. Further research is warranted to elucidate the therapeutic potential of metformin through these mechanisms.
Keywords: Metformin, Breast Cancer, Mir-125A, Methylation, HER2, DNMT1, Epigenetics -
Pages 286-302
Esophageal cancer presents a challenge in gastroenterology and traditional chemotherapy and radiation therapy have less therapeutic activity with severe side effects. Thus, there is need for effective and safer alternatives. Probiotics, particularly Lactobacillus plantarum (L. plantarum) and its bacteriocins, might prevent or treat esophageal tumors. We aimed to investigate the use of L. plantarum and its bacteriocin as esophageal cancer therapy. First, we obtained 100 isolates of Lactobacillus spp. from dairy product samples. They screened for bacteriocin production and identified by PCR and gel electrophoresis for 16S ribosomal RNA gene. Bacteriocin was partially purified and tested against two different pathogens. Both L. plantarum and its bacteriocin were examined for cytotoxicity in vitro against esophageal cancer cell line (SK-GT4) and normal rat embryo fibroblast (REF) cells by MTT assay. Apoptosis was determined using an acridine orange /propidium iodide assay. The results showed that the isolate gives a high bacteriocin production about (2000AU/ml). In addition to antimicrobial activity, there was significant anticancer activity. L. plantarum had an IC50 of 51.01 CFU/ml and bacteriocin IC50 of 281.9 AU/ml against cancer cells. Both showed no cytotoxicity towards normal REF cells. Furthermore, there was a significant increase in apoptosis induction and in caspase-3 activity in cancer cells treated with L. plantarum and bacteriocin compared to untreated cells. In conclusion, L. plantarum and its bacteriocin show potent killing effect against esophageal cancer cells with no effect against normal cells indicating safety and selectivity with activation of apoptosis via caspase-3 induction suggesting potential clinical advantage.
Keywords: Lactobacillus Plantarum, Bacteriocin, Antitumor Activity, Apoptosis, Gastro-Intestinal Tumors -
Pages 303-324
Liver cancer treatment faces significant obstacles such as resistance, recurrence, metastasis, and toxicity to healthy cells. Biometallic nanoparticles (NPs) have emerged as a promising approach to address these challenges. In this study, copper oxide-silver (Ag-doped CuO) NPs were prepared using a reduction method with Ephedra intermedia extract. The physicochemical properties of the NPs were evaluated using various techniques such as Field emission scanning electron microscopy (FESEM), Transmission Electron Microscope (TEM), X-ray diffraction (XRD), and Fourier-transform infrared spectroscopy (FTIR). Additionally, this study has evaluated nitric oxide levels (NO), reactive oxygen species (ROS) production, Bax, Bcl2, P53, and Caspase3 genes expression, as well as cell viability within 24 hours in liver cancer cell line HepG2. FESEM and TEM imaging confirmed the nanostructural nature of the synthesized particles with sizes ranging from 31.27 to 88.98 nanometers. XRD analysis confirmed the crystal structure of the NPs. Comparative analysis showed that the IC50 values of the Ag-doped CuO NPs were significantly lower than that of the plant extracts. Molecular studies showed significantly increased expression of Bax, Caspase3, and P53 genes, inducing apoptosis in cancer cells, and downregulation of Bcl2 as a pro-metastasis gene. Additionally, the presence of Ag-doped CuO NPs significantly increased NO activity enzyme and ROS generation compared to the plant extract. The biosynthesized Ag-doped CuO NPs demonstrated the ability to induce apoptosis, increase ROS production, and enhance NO enzyme activity in HepG2 cancer cells, suggesting their potential as a therapeutic agent for liver cancer.
Keywords: Liver Cancer, Apoptosis, Silver Nanoparticles, Copper Nanoparticles, Ephedra Intermedia Extract, Anticancer Properties -
Pages 325-336
Coronary artery diseases (CAD) represent a significant global health concern and are recognized as a primary contributor to mortality on a worldwide scale. Early diagnosis of CAD is one of promising goal to manage this disorder. Recent investigations have highlighted the pivotal involvement of microRNAs (miRNAs) in diverse health conditions, notably CAD. The principal objective of this investigation was to identify appropriate miRNAs that could be employed for the early detection of CAD. In the present study, we analyzed dataset of CAD (GSE113079) and 100 differentially expressed mRNAs (DEmRNAs) were detected. The miRNAs that have a significant interaction with DEmRNAs were chosen. By computational prediction method, 5 miRNAs (miR-106b-5p, miR-20a-3p, miR-17-3p, miR-146a-5p, and miR-155-3p) were selected. Finally, we assessed the anticipated expression levels of microRNAs in CAD patients and healthy control groups. Our findings revealed a statistically significant elevation solely in the expression level of miR-106b-5p within the CAD group when compared to the control group (p>0.001). Our study demonstrated an elevation in the expression of miR-106b-5p in individuals diagnosed with CAD. This microRNA may be used as a diagnostic biomarker in patients with CAD. However, further investigations are needed to confirm these results.
Keywords: Circulating Mirna, Coronary Artery Disease, Mirna-Mrna Intraction, Biomarker