Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.246
Filter
1.
World J Urol ; 42(1): 468, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110253

ABSTRACT

INTRODUCTION: Bladder cancer (BC) is a prevalent malignancy with high recurrence rates. Patient-derived bladder cancer organoids (BCO) pose as a promising approach in both, disease modeling and individualized treatment screening. The aim of this study was to investigate the transcriptomic plasticity in BCOs as a function of cultivation times to define ideal time periods for the applications envisioned. METHODS: Tumor samples of three patients with pathologically confirmed non-muscle invasive and muscle-invasive bladder cancer were included in this study and expanded as BCOs. RNA expression was investigated at different time periods of cells in culture using differential gene expression for overall transcript expression and quantitative real-time PCR (qRT-PCR) for pathological relevant markers. RESULTS: Differential gene expression of the BCO lines was investigated across passages 1-4, in passages 5-9 and above 9, respectively. Analysis of the entire transcriptome of the respective BCO lines revealed consistent profiles without significant alterations throughout the cultivation and expansion procedure. Notably, key transcripts like TP53, PIK3CA, BRCA1, among others, exhibited stable expression levels in the quantitative RNA analysis during the cultivation period. CONCLUSION: The robust transcriptome during BCO cultivation advocates for the use of earlier passages of BCOs in personalized medicine providing a time-efficient drug screening option to accelerate the counseling of patients' treatment options. Higher passages of BCOs still hold the potential in topics demanding for expanded cell masses such as medical device development and others.


Subject(s)
Organoids , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Organoids/metabolism , Gene Expression Regulation, Neoplastic , Male , Transcriptome , Tumor Cells, Cultured , Female
2.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126111

ABSTRACT

Curcumin (Cur) is a heavily used complementary derived drug from cancer patients. Spheroid samples derived from 82 patients were prepared and treated after 48 h with two Cur formulations (CurA, CurB) in mono- and combination therapy. After 72 h, cell viability and morphology were assessed. The Cur formulations had significant inhibitory effects of -8.47% (p < 0.001), CurA of -10.01% (-50.14-23.11%, p = 0.001) and CurB of -6.30% (-33.50-19.30%, p = 0.006), compared to their solvent controls Polyethylene-glycol, ß-Cyclodextrin (CurA) and Kolliphor-ELP, Citrate (CurB). Cur formulations were more effective in prostate cancer (-19.54%) and less effective in gynecological non-breast cancers (0.30%). CurA showed better responses in samples of patients <40 (-13.81%) and >70 years of age (-17.74%). CurB had stronger effects in metastasized and heavily pretreated tumors. Combinations of Cur formulations and standard therapies were superior in 20/47 samples (42.55%) and inferior in 7/47 (14.89%). CurB stimulated chemo-doublets more strongly than monotherapies (-0.53% vs. -6.51%, p = 0.022) and more effectively than CurA (-6.51% vs. 3.33%, p = 0.005). Combinations of Cur formulations with Artesunate, Resveratrol and vitamin C were superior in 35/70 (50.00%) and inferior in 16/70 (22.86%) of samples. Cur formulations were significantly enhanced by combination with Artesunate (p = 0.020). Cur formulations showed a high variance in their anti-cancer effects, suggesting a need for individual testing before administration.


Subject(s)
Antineoplastic Agents , Curcumin , Spheroids, Cellular , Humans , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/administration & dosage , Female , Aged , Male , Middle Aged , Spheroids, Cellular/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Adult , Cell Survival/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Drug Compounding , Tumor Cells, Cultured
3.
Exp Cell Res ; 441(1): 114155, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39002689

ABSTRACT

At least one-third of patients with epithelial ovarian cancer (OC) present ascites at diagnosis and almost all have ascites at recurrence especially because of the propensity of the OC cells to spread in the abdominal cavity leading to peritoneal metastasis. The influence of ascites on the development of pre-metastatic niches, and on the biological mechanisms leading to cancer cell colonization of the mesothelium, remains poorly understood. Here, we show that ascites weakens the mesothelium by affecting the morphology of mesothelial cells and by destabilizing their distribution in the cell cycle. Ascites also causes destabilization of the integrity of mesothelium by modifying the organization of cell junctions, but it does not affect the synthesis of N-cadherin and ZO-1 by mesothelial cells. Moreover, ascites induces disorganization of focal contacts and causes actin cytoskeletal reorganization potentially dependent on the activity of Rac1. Ascites allows the densification and reorganization of ECM proteins of the mesothelium, especially fibrinogen/fibrin, and indicates that it is a source of the fibrinogen and fibrin surrounding OC spheroids. The fibrin in ascites leads to the adhesion of OC spheroids to the mesothelium, and ascites promotes their disaggregation followed by the clearance of mesothelial cells. Both αV and α5ß1 integrins are involved. In conclusion ascites and its fibrinogen/fibrin composition affects the integrity of the mesothelium and promotes the integrin-dependent implantation of OC spheroids in the mesothelium.


Subject(s)
Ascites , Fibrin , Fibrinogen , Integrin alpha5beta1 , Ovarian Neoplasms , Spheroids, Cellular , Tumor Microenvironment , Humans , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ascites/pathology , Ascites/metabolism , Integrin alpha5beta1/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Fibrinogen/metabolism , Fibrin/metabolism , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Receptors, Vitronectin/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Adhesion , Peritoneum/pathology , Peritoneum/metabolism , Epithelium/metabolism , Epithelium/pathology , Cadherins/metabolism , Tumor Cells, Cultured
4.
Ann Anat ; 255: 152298, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971450

ABSTRACT

Cancer research involves significant animal consumption and suffering. Tumor cells can be differentiated in vitro into three-dimensional organoids that resemble the primary tumor. In basic cancer research, however, tumor organoids are usually only used alongside animal experiments. We have established an easy-to-perform protocol that allows to culture KRAS-driven lung tumor cells as organoids for extended periods of time. Like the corresponding tumors in mice, the organoids produce surfactant protein C but no markers of airway epithelial cells (e.g. SCGB1A1, KRT5). The organoids can be passaged as single cell suspensions. Our organoid model contributes to replace animal experiments with cell culture systems and can be used for drug testing or functional studies in cancer research.


Subject(s)
Lung Neoplasms , Organoids , Animals , Organoids/pathology , Mice , Lung Neoplasms/pathology , Cell Culture Techniques/methods , Tumor Cells, Cultured
5.
Asian Pac J Cancer Prev ; 25(7): 2371-2379, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39068570

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) arises from hepatocytes and accounts for 90% of primary liver cancer. Reasons for HCC prognosis remaining dismal are that HCC is asymptomatic in its early stages, leading to late diagnosis, and it is markedly resistant to conventional chemo- and radiotherapy. In this study, we investigated RNA interference (RNAi)-based treatment for HCC by targeting MDK. AIM: The present study aimed to evaluate MDK serum levels as a diagnostic biomarker for HCC detection and the effect of MDK silencing by RNAi on HCC. SUBJECTS AND METHODS: A total of 140 participants, including 120 patients diagnosed with HCC and 20 healthy volunteers were enrolled in this study, all patients who underwent liver resection were sampled for tumor and adjacent non-tumor liver tissues, in addition to 5 ml of blood sample. Midkine expression levels were evaluated by ELISA and by qRT-PCR. The in vitro transfection and gene knockdown efficiency of midkine by MDK-siRNA was detected by qRT-PCR and ELISA. Gene knockdown effect at the molecule level on the proliferation of HepG2 in vitro was determined by cell counting. RESULTS: The results showed that the expression of MDK was significantly increased in the serum of HCC patients compared to control serum samples with P<0.001 and significant elevated expression levels of MDK in tumor tissues compared to non-tumor ones with P<0.001. It also showed that down-regulation of MDK using RNAi can significantly inhibit HepG2 cells. CONCLUSION: Molecular targeting of MDK using RNAi interference decreases proliferation and could be a therapeutic target.


Subject(s)
Biomarkers, Tumor , Carcinoma, Hepatocellular , Cell Proliferation , Liver Neoplasms , Midkine , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Midkine/genetics , Male , Female , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Prognosis , RNA Interference , Genetic Therapy/methods , Case-Control Studies , RNA, Small Interfering/genetics , Hep G2 Cells , Follow-Up Studies , Adult , Tumor Cells, Cultured
6.
Asian Pac J Cancer Prev ; 25(7): 2391-2396, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39068572

ABSTRACT

OBJECTIVE: Now a day's cancerous diseases are the most prevalent life threatening that spreading because of the lifestyle. Its due to uncontrolled growth of cell which can be cured if diagnosed in early stage. Treatment of cancer depends on the various internal and external factors causing cancer. The main objective of this study is using herbal based medicine to manage breast cancer, the second most common type of cancer in the world. METHODS: In this study, the anticancer effect of two Iraqi date palm part extracts (leaves and fruits) against panel of breast cancer cell lines (AMJ13و MCF7, MDA-MB-231, CAL51) in vitro to evaluate their possible antitumor effect and their safety on normal cell line (MEF). RESULTS: The Phoenix dactylifera (dray Zahdi) fresh leave extract showed highly cytotoxic effects in all breast cancer cell lines. The leaves extract was showed concentration dependent cytotoxicity effects after 72 h exposure time. Leave extracts was effective against AMJ13 cell line. The effective concentrations in both cancer cells ranged from 2500-20000 µg/ ml with inhibition percentage against AMJ13 was (66.7, 70.6, 53, and 54%). While the effect against MCF7, MDA-MB, and CAL51 cell lines were less with significant effect only at two concentrations (10000- 20000 µg/ ml) causing 64.3, and 64.3% growth inhibition respectively in MCF7, and 40, and 50% respectively in MDA-MB, and 44.0, and 52.0% respectively in CAL51. The dray date fruit extract has no significant cytotoxicity against all the cancer cells. Both extracts have no effect against normal fibroblast cells. CONCLUSION: In conclusion, Phoenix dactylifera fresh leave extract shows promising anticancer properties while the fruit extract has no direct anticancer effect.


Subject(s)
Breast Neoplasms , Cell Proliferation , Fruit , Phoeniceae , Plant Extracts , Plant Leaves , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Phoeniceae/chemistry , Plant Extracts/pharmacology , Fruit/chemistry , Plant Leaves/chemistry , Female , Cell Proliferation/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Tumor Cells, Cultured
7.
Asian Pac J Cancer Prev ; 25(7): 2539-2550, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39068589

ABSTRACT

OBJECTIVE: Colorectal cancer is a significant global health concern with high mortality rates. Silibinin is a compound derived from milk thistle with anticancer properties and may be a potential treatment option for colorectal cancer. Its poor solubility limits its clinical application, but various strategies, such as nanoparticle encapsulation, have shown promise. In this study, a PEGylated niosomal drug delivery system was used to enhance the solubility of silibinin, and its anti-proliferative effects were evaluated against human colorectal cancer cell lines. METHODS: The silibinin-loaded PEGylated niosomal nanoparticles (NIO-SIL) were fabricated using the thin-film hydration method and characterized with dialysis bag, AFM, SEM, DLS, and FTIR systems. Finally, the cancerous cells and human normal cells were treated with NIO-SIL and pure silibinin. The proliferation, apoptosis, and cell cycle of these cells were evaluated. Subsequently, the expression of Bax, Bcl-2, p53, and cyclin D1 genes was measured using real-time PCR. RESULT: The drug release profile, size, morphology, and chemical interactions of the synthesized PEGylated niosomal nanoparticles were suitable for use as a drug delivery system. Both pure silibinin and NIO-SIL could reduce the proliferation of cancerous cells, induce apoptosis, and cause cell cycle arrest, with no significant negative effects reported on human normal cells. Both pure silibinin and NIO-SIL reduced the expression of the Bcl-2 and cyclin D1 genes while increasing the expression of Bax and p53. (p-value < 0.05 *). CONCLUSION: The outcomes of this study indicate the high potential of PEGylated niosomal nanoparticles for encapsulation and delivery of silibinin to cancer cells, with no negative effects on normal cells.


Subject(s)
Apoptosis , Cell Proliferation , Nanoparticles , Polyethylene Glycols , Silybin , Humans , Silybin/pharmacology , Silybin/chemistry , Apoptosis/drug effects , Nanoparticles/chemistry , Cell Proliferation/drug effects , Polyethylene Glycols/chemistry , Liposomes/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cell Cycle/drug effects , Drug Delivery Systems/methods , Tumor Cells, Cultured , Cell Line, Tumor
8.
Drug Des Devel Ther ; 18: 3157-3173, 2024.
Article in English | MEDLINE | ID: mdl-39071813

ABSTRACT

Background: Tumor-Node-Metastasis (TNM) stage of gastric cancer (GC) is one of the main factors affecting clinical outcome. The aim of this study was to explore the targets related to TNM stage of GC, and screening natural bioactive drug. Methods: RNA sequencing data of the TCGA-STAD cohort were downloaded from UCSC database. Genes associated with TNM staging were identified by weighted gene co-expression network analysis (WGCNA). Univariate Cox regression, least absolute shrinkage and selection operator (LASSO), extreme gradient boosting (Xgboost), random forest (RF) and cytohubba plug-in of cytoscope were applied to screen hub genes. Natural bioactive ingredients were available from the HERB database. Molecular docking was used to evaluate the binding activity of active ingredients to the hub protein. CCK-8, flow cytometry, transwell and Western blot assays were used to analyze the effects of diosgenin on GC cells. Results: 898 TNM-related genes were screened out through WGCNA. Three genes associated with GC progression/prognosis were identified, including nuclear receptor subfamily 3 group C member 2 (NR3C2), solute carrier family 1 member 5 (SLC1A5) and FAT atypical cadherin 1 (FAT1) based on the machine learning algorithms and hub co-expression network analysis. Diosgenin had good binding activity with SLC1A5. SLC1A5 was highly expressed in GC and was closely associated with tumor stage, overall survival and immune infiltration of GC patients. Diosgenin could inhibit cell viability and invasive ability, promote apoptosis and induce cell cycle arrest in G0/G1 phase. In addition, diosgenin promoted cleaved caspase 3 expression and inhibited Ki67, cyclin D1, p-S6K1, and SLC1A5 expression levels, while the mTORC1 activator (MHY1485) reversed this phenomenon. Conclusion: For the first time, this work reports diosgenin may inhibit the activation of mTORC1 signaling through targeting SLC1A5, thereby inhibiting the malignant behaviors of GC cells.


Subject(s)
Cell Proliferation , Diosgenin , Mechanistic Target of Rapamycin Complex 1 , Molecular Docking Simulation , Stomach Neoplasms , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , Cell Proliferation/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Diosgenin/pharmacology , Diosgenin/chemistry , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Disease Progression , Drug Screening Assays, Antitumor , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Dose-Response Relationship, Drug , Tumor Cells, Cultured
9.
World J Surg Oncol ; 22(1): 193, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39054546

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the most common cancers worldwide. Tumor microenvironment plays an important role in tumor progression. This study aims to explore the role of cancer-associated fibroblasts (CAFs) in GC and the underlying mechanism. METHODS: Cell viability, proliferation, invasion and migration were assessed by MTT, EdU, transwell and wound healing assays, respectively. Sphere formation assay was used to evaluate cell stemness. Glucose consumption, lactate production and ATP consumption were measured to assess glycolysis. In addition, The RNA and protein expression were detected by qRT-PCR and western blot. The interaction between wingless Type MMTV Integration Site Family, Member 5 A (WNT5A) and hexokinase 2 (HK2) was verified by Co-immunoprecipitation. The xenograft model was established to explore the function of CAFs on GC tumor growth in vivo. RESULTS: CAFs promoted the proliferation, metastasis, stemness and glycolysis of GC cells. WNT5A was upregulated in CAFs, and CAFs enhanced WNT5A expression in GC cells. Knockdown of WNT5A in either GC cells or CAFs repressed the progression of GC cells. In addition, WNT5A promoted HK2 expression, and overexpression of HK2 reversed the effect of WNT5A knockdown in CAFs on GC cells. Besides, knockdown of WNT5A in CAFs inhibits tumor growth in vivo. CONCLUSION: CAF-derived WNT5A facilitates the progression of GC via regulating HK2 expression.


Subject(s)
Cancer-Associated Fibroblasts , Cell Movement , Cell Proliferation , Glycolysis , Hexokinase , Stomach Neoplasms , Wnt-5a Protein , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Humans , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Animals , Mice , Hexokinase/metabolism , Hexokinase/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Mice, Nude , Tumor Microenvironment , Xenograft Model Antitumor Assays , Prognosis , Mice, Inbred BALB C , Apoptosis , Male , Cell Line, Tumor
10.
Oncoimmunology ; 13(1): 2373519, 2024.
Article in English | MEDLINE | ID: mdl-38988823

ABSTRACT

Biomarkers for cancer immunotherapy are an unmet medical need. The group of Daniela Thommen at the NKI recently reported on novel methodologies based on short-term cultures of patient-derived tumor fragments whose cytokine concentrations in the supernatants and activation markers on infiltrating T cells were associated with clinical response to PD-1 blockade. We set up a similar culture technology with tumor-derived fragments using mouse tumors transplanted into syngeneic immunocompetent mice to test an agonist anti-CD137 mAb and its combinations with anti-PD-1 and/or anti-TGF-ß. Increases in IFNγ concentrations in the tissue culture supernatants were detected upon in-culture activation with the anti-CD137 and anti-PD-1 mAb combinations or concanavalin A as a positive control. No other cytokine from a wide array was informative of stimulation with these mAbs. Interestingly, increases in Ki67 and other activation markers were substantiated in lymphocytes from cell suspensions gathered at the end of 72 h cultures. In mice bearing bilateral tumors in which one was excised prior to in vivo anti-CD137 + anti-PD-1 treatment to perform the fragment culture evaluation, no association was found between IFNγ production from the fragments and the in vivo therapeutic outcome in the non-resected contralateral tumors. The experimental system permitted freezing and thawing of the fragments with similar functional outcomes. Using a series of patient-derived tumor fragments from excised solid malignancies, we showed IFNγ production in a fraction of the studied cases, that was conserved in frozen/thawed fragments. The small tumor fragment culture technique seems suitable to preclinically explore immunotherapy combinations.


Subject(s)
Immunotherapy , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Mice , Humans , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Female , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/pathology , Interferon-gamma/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Tumor Cells, Cultured , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
11.
Drug Des Devel Ther ; 18: 1833-1853, 2024.
Article in English | MEDLINE | ID: mdl-38828018

ABSTRACT

Purpose: Given the potent immunostimulatory effects of bacterial outer membrane vesicles (OMVs) and the significant anti-colon tumor properties of Parabacteroides distasonis (Pd), this study aimed to elucidate the role and potential mechanisms of Pd-derived OMVs (Pd-OMVs) against colon cancer. Methods: This study isolated and purified Pd-OMVs from Pd cultures and assessed their characteristics. The effects of Pd-OMVs on CT26 cell uptake, proliferation, and invasion were investigated in vitro. In vivo, a CT26 colon tumor model was used to investigate the anti-colon tumor effects and underlying mechanisms of Pd-OMVs. Finally, we evaluated the biosafety of Pd-OMVs. Results: Purified Pd-OMVs had a uniform cup-shaped structure with an average size of 165.5 nm and a zeta potential of approximately -9.56 mV, and their proteins were associated with pathways related to immunity and apoptosis. In vitro experiments demonstrated that CT26 cells internalized the Pd-OMVs, resulting in a significant decrease in their proliferation and invasion abilities. Further in vivo studies confirmed the accumulation of Pd-OMVs in tumor tissues, which significantly inhibited the growth of colon tumors. Mechanistically, Pd-OMVs increased the expression of CXCL10, promoting infiltration of CD8+ T cells into tumor tissues and expression of pro-inflammatory factors TNF-α, IL-1ß, and IL-6. Notably, Pd-OMVs demonstrated a high level of biosafety. Conclusion: This paper elucidates that Pd-OMVs can exert significant anti-colon tumor effects by upregulating the expression of the chemokine CXCL10, thereby increasing the infiltration of CD8+ T cells into tumors and enhancing antitumor immune responses. This suggests that Pd-OMVs may be developed as a novel nanoscale potent immunostimulant with great potential for application in tumor immunotherapy. As well as developed as a novel nano-delivery carrier for combination with other antitumor drugs.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Proliferation , Chemokine CXCL10 , Colonic Neoplasms , Mice, Inbred BALB C , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/drug therapy , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Mice , Cell Proliferation/drug effects , Chemokine CXCL10/metabolism , Chemokine CXCL10/immunology , Bacterial Outer Membrane/immunology , Bacterial Outer Membrane/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Humans , Neoplasms, Experimental/pathology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/drug therapy , Drug Screening Assays, Antitumor , Tumor Cells, Cultured
12.
World J Surg Oncol ; 22(1): 152, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849867

ABSTRACT

BACKGROUND: Although sorafenib has been consistently used as a first-line treatment for advanced hepatocellular carcinoma (HCC), most patients will develop resistance, and the mechanism of resistance to sorafenib needs further study. METHODS: Using KAS-seq technology, we obtained the ssDNA profiles within the whole genome range of SMMC-7721 cells treated with sorafenib for differential analysis. We then intersected the differential genes obtained from the analysis of hepatocellular carcinoma patients in GSE109211 who were ineffective and effective with sorafenib treatment, constructed a PPI network, and obtained hub genes. We then analyzed the relationship between the expression of these genes and the prognosis of hepatocellular carcinoma patients. RESULTS: In this study, we identified 7 hub ERGs (ACTB, CFL1, ACTG1, ACTN1, WDR1, TAGLN2, HSPA8) related to drug resistance, and these genes are associated with the cytoskeleton. CONCLUSIONS: The cytoskeleton is associated with sorafenib resistance in hepatocellular carcinoma. Using KAS-seq to analyze the early changes in tumor cells treated with drugs is feasible for studying the drug resistance of tumors, which provides reference significance for future research.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Drug Resistance, Neoplasm , Liver Neoplasms , Sorafenib , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Humans , Sorafenib/pharmacology , Sorafenib/therapeutic use , Liver Neoplasms/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Prognosis , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Cytoskeleton/drug effects , Cytoskeleton/pathology , Cytoskeleton/metabolism , Biomarkers, Tumor/genetics , Tumor Cells, Cultured , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Profiling
13.
Asian Pac J Cancer Prev ; 25(6): 2133-2138, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38918676

ABSTRACT

OBJECTIVE: This study aimed to discover the cytotoxic effect of YH239-EE and YH239 alone and their enantiomer potency in cytotoxic effect on the MCF7 cell line. METHODS: We used the cytotoxic study on MDM2 cell lines by detecting the percentage of apoptosis and necrosis by annexin v methods. RESULT: This result shows that YH239-EE causes more apoptosis and necrosis 40% in comparison to YH239 without ethyl ester, about 4.92 %,  and The (+) enantiomer of YH239-EE demonstrated a markedly higher induction of apoptosis and necrosis (84.48%) in MCF7 cells compared to the (-) enantiomer (48.71%). CONCLUSION: The ethyl ester group in YH239-EE might play a crucial role in enhancing the compound's ability to induce cell death, and The high efficacy of the (+) enantiomer of YH239-EE in inducing cell death in MCF7 cells suggests it may be a more promising therapeutic candidate for breast cancer treatment, specifically for subtypes represented by MCF7 cells.


Subject(s)
Apoptosis , Breast Neoplasms , Humans , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , MCF-7 Cells , Female , Cell Proliferation/drug effects , Tumor Cells, Cultured , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Stereoisomerism
14.
Asian Pac J Cancer Prev ; 25(6): 2147-2157, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38918678

ABSTRACT

OBJECTIVE: Lung cancer is the most common malignancy and among the leading cause of cancer death worldwide. Therefore, there is an important need for biomarkers that can be used in the early diagnosis of the disease and in the follow-up of treatment. Circular RNAs (circRNAs) have a covalently closed circular structure that lacks 3' and 5' polar ends and is resistant to RNAase enzymes. Due to these properties, they can be stably found in body fluids. Therefore, they can serve as potential biomarkers in the diagnosis, monitoring of therapeutic response and prognosis of cancer. In our study, we aimed to investigate the expression levels of circRNA molecules in the treatment of lung cancer and to determine those that have the potential to be biomarkers. METHODS: In this in vitro study, expression levels of 163 circRNAs were investigated in A549 cells, a non-small cell lung cancer cell line, before and after treatment with carboplatin and pemetrexed. Total RNA isolation and cDNA synthesis were performed after treatments. Expression levels of circRNA genes were determined by RT-qPCR method with the designed divergent primer sequences. RESULTS: The study revealed the characterisation of differentially expressed circRNAs by treatment in lung cancer cells. Of them, hsa_circ_0001320 is not expressed in cancer cells, is expressed only after treatment, and increased the level of its expression in response to combination therapy. CONCLUSION: As a result, while carboplatin, pemetrexed, and combined drug applications changed the expression levels of some circRNAs in lung cancer cells, some circRNAs were expressed only after treatment. In treatment follow-up and management, hsa_circ_0001320 has been identified as potential biomarker candidate.


Subject(s)
Biomarkers, Tumor , Lung Neoplasms , RNA, Circular , Humans , RNA, Circular/genetics , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Biomarkers, Tumor/genetics , Prognosis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carboplatin/therapeutic use , Pemetrexed/therapeutic use , Gene Expression Regulation, Neoplastic , Tumor Cells, Cultured
15.
Asian Pac J Cancer Prev ; 25(6): 2169-2176, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38918680

ABSTRACT

BACKGROUND: Tongue cancer is the most prevalent type of oral cancer. Recently, natural compounds have been considered important resources for several anticancer drugs. Thymoquinone (TQ) exhibits a potent anti-cancer effect. 5-Fluorouracil (5-FU) is a chemotherapeutic drug that has been utilized in the treatment of cancer. Recently, combination therapy has gained popularity as a treatment option for patients with cancer. OBJECTIVES: The present study was carried out to assess the cytotoxic effect of 5-Fluorouracil (5-FU), Thymoquinone (TQ), and their combination on tongue squamous cell carcinoma cell line (HNO-97). METHODS: Tongue carcinoma cell line (HNO-97) was maintained in cultured flasks and the cells were divided into four groups; group Ι: control untreated group, group ΙΙ: HNO-97-treated cells with different concentrations of 5-FU from 0.5 µM/ml to 3µM/ml, group ΙIΙ: HNO-97-treated cells with different concentrations of TQ from 7.25µM/ml to 23.05µM/ml, and group ΙV: HNO-97-treated cells with both 5-FU and TQ in serial concentrations  till (IC50) in a dose of 27.44 µM/ml. Determination of the cytotoxic effect of the tested agents on the HNO-97 cell line was done using methyl thiazole tetrazolium assay, nuclear morphometric analysis, microscopic examination, and annexin-v/ propidium iodide staining assay. RESULT: The findings revealed that the cytotoxic effect of 5-FU, TQ, and their combination on tongue squamous cell carcinoma cell line (HNO-97) was dose-dependent. The microscopic examination revealed that 5-FU, TQ alone, or their combination induced apoptotic cell death. P-value < 0.05 was statistically significant. CONCLUSION: The combination of 5-FU and TQ produced a marked cytotoxic effect on HNO-97 cells.


Subject(s)
Apoptosis , Benzoquinones , Carcinoma, Squamous Cell , Cell Proliferation , Fluorouracil , Tongue Neoplasms , Humans , Fluorouracil/pharmacology , Benzoquinones/pharmacology , Tongue Neoplasms/drug therapy , Tongue Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Apoptosis/drug effects , Cell Proliferation/drug effects , Tumor Cells, Cultured , Antineoplastic Combined Chemotherapy Protocols/pharmacology , In Vitro Techniques , Cell Line, Tumor , Drug Synergism
16.
STAR Protoc ; 5(2): 103103, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38829737

ABSTRACT

T cell acute lymphoblastic leukemia (T-ALL) is a rare but aggressive hematological cancer that occurs primarily in children and adolescents. Here, we present a protocol for in vitro co-culture assay that enables robust expansion of primary T-ALL cells. We describe steps for seeding T-ALL and stromal cells in 3D organoids and subsequent flow analysis to capture the T-ALL cell growth for long-term culture. This protocol provides a valuable platform for in vitro functional studies and drug screenings using patient-derived cells. For complete details on the use and execution of this protocol, please refer to Rivera et al.1.


Subject(s)
Coculture Techniques , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Coculture Techniques/methods , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Organoids/pathology , Organoids/cytology , Cell Proliferation , Cell Culture Techniques/methods , Tumor Cells, Cultured
17.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38928438

ABSTRACT

Anaplastic thyroid cancer (ATC) is one of the deadliest human cancers and represents <2% of thyroid carcinomas. A therapeutic target for ATC is represented by anaplastic lymphoma kinase (ALK) rearrangements, involved in tumor growth. Crizotinib is an oral small-molecule tyrosine kinase inhibitor of the ALK, MET, and ROS1 kinases, approved in ALK-positive non-small cell lung cancer. Until now, the effect of crizotinib in "primary human ATC cells" (pATCs) with transforming striatin (STRN)-ALK fusion has not been reported in the literature. In this study, we aimed to obtain pATCs with STRN-ALK in vitro and evaluate the in vitro antineoplastic action of crizotinib. Thyroid surgical samples were obtained from 12 ATC patients and 6 controls (who had undergone parathyroidectomy). A total of 10/12 pATC cultures were obtained, 2 of which with transforming STRN-ALK fusion (17%). Crizotinib inhibited proliferation, migration, and invasion and increased apoptosis in 3/10 pATC cultures (2 of which with/1 without STRN-ALK), particularly in those with STRN-ALK. Moreover, crizotinib significantly inhibited the proliferation of AF cells (a continuous cell line obtained from primary ATC cells). In conclusion, the antineoplastic activity of crizotinib has been shown in human pATCs (with STRN-ALK) in preclinical studies in vitro, opening the way to future clinical evaluation in these patients.


Subject(s)
Anaplastic Lymphoma Kinase , Apoptosis , Cell Proliferation , Crizotinib , Protein Kinase Inhibitors , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Humans , Crizotinib/pharmacology , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Carcinoma, Anaplastic/pathology , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , Cell Proliferation/drug effects , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Male , Female , Antineoplastic Agents/pharmacology , Middle Aged , Cell Movement/drug effects , Aged , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Tumor Cells, Cultured , Cell Line, Tumor , Calmodulin-Binding Proteins , Membrane Proteins , Nerve Tissue Proteins
18.
Radiat Oncol ; 19(1): 84, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926729

ABSTRACT

BACKGROUND: At present, it has been found that many patients have acquired resistance to radiotherapy, which greatly reduces the effect of radiotherapy and further affects the prognosis. CircRNAs is involved in the regulation of radiosensitivity of many kinds of tumor cells. Therefore, the main purpose of this study is to explore the regulatory effect of CircRNA_101491 on radiosensitivity of ESCC and its related mechanism. METHODS: We established ESCC radiation-resistant cell line (KYSE150R cell) by gradient dose method, and tested the difference of KYSE150 between KYSE150R cell and parent cell in vitro. Then, after knocking down the expression of CircRNA_101491, a series of in vitro experiments were conducted to verify the effects of CircRNA_101491 on the phenotype and radiosensitivity of KYSE150R cells, and further analyzed the related regulatory mechanism. In addition, we also used the model of transplanted tumor in nude mice to investigate the effect of CircRNA_101491 on the radiosensitivity of ESCC in vivo. RESULTS: According to a series of in vitro experiments, we confirmed that KYSE150R cells lost the epithelial phenotype and obtained interstitial cell-like phenotype, and found that CircRNA_101491 was highly expressed in KYSE150R cells. In addition, we found that knocking down the expression of CircRNA_101491 will lift the inhibition of miR-125a-5p, and then reverse the process of EMT, accelerate the process of apoptosis, thus play a role in radiosensitization. The in vivo experiment of transplanted tumor in nude mice also showed that knocking down the expression of CircRNA_101491 could enhance the radiosensitivity of ESCC. CONCLUSION: In conclusion, we confirmed that interfering with the expression of CircRNA_101491 can relieve the inhibition of miR-125a-5p, thus reverse the process of interstitial phenotype, accelerate the process of apoptosis, and enhance the radiosensitivity of ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Mice, Nude , MicroRNAs , RNA, Circular , Radiation Tolerance , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , RNA, Circular/genetics , Humans , Mice , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/radiotherapy , Esophageal Squamous Cell Carcinoma/metabolism , Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Cell Line, Tumor , Xenograft Model Antitumor Assays , Tumor Cells, Cultured
19.
J Enzyme Inhib Med Chem ; 39(1): 2339901, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38864175

ABSTRACT

The spices and aromatic herbs were used not only in cooking to add flavour and smell to dishes but also for medicinal use. Nigella sativa, also called black cumin, is one of the species that contains an important bioactive component, thymoquinone (TQ), which has antioxidant, anti-inflammatory, antimicrobial, and antidiabetic effects. Curcuma longa, which also includes curcumin, has numerous anti-cancer properties. However, the bioavailability of curcumin is lower than that of its analogs. An analog of curcumin (EF-24), which has better bioavailability than curcumin, is capable of exerting a high anti-cancer effect. In our study, we determined the effects of PON1 enzyme activity on the proliferation and aggressiveness of glioblastoma cancer treated with TQ and EF-24 from lysates of the glioblastoma cell line U87MG. The results were determined as increased PON1 activity after treatment with TQ and EF-24 in the U87MG cell line (p < 0.0001).


Subject(s)
Aryldialkylphosphatase , Benzoquinones , Cell Proliferation , Curcumin , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioblastoma , Humans , Aryldialkylphosphatase/metabolism , Aryldialkylphosphatase/antagonists & inhibitors , Glioblastoma/drug therapy , Glioblastoma/pathology , Benzoquinones/pharmacology , Benzoquinones/chemistry , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/chemical synthesis , Cell Proliferation/drug effects , Molecular Structure , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Tumor Cells, Cultured
20.
Leukemia ; 38(8): 1742-1750, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906964

ABSTRACT

Activated B-cell-like diffuse large B-cell lymphoma (ABC-DLBCL) is driven by aberrant activation of the B-cell receptor (BCR) and the TLR/MyD88 signaling pathways. The heat-shock protein HSP110 is a candidate for their regulation as it stabilizes MyD88. However, its role in overall BCR signaling remains unknown. Here, we used first-in-class HSP110 inhibitors to address this question. HSP110 inhibitors decreased the survival of several ABC-DLBCL cell lines in vitro and in vivo, and reduced the phosphorylation of BCR signaling kinases, including BTK and SYK. We identified an interaction between HSP110 and SYK and demonstrated that HSP110 promotes SYK phosphorylation. Finally, the combination of the HSP110 inhibitor with the PI3K inhibitor copanlisib decreases SYK/BTK and AKT phosphorylation synergistically, leading to suppression of tumor growth in cell line xenografts and strong reduction in patient-derived xenografts. In conclusion, by regulating the BCR/TLR signaling pathway, HSP110 inhibitors are potential drug candidates for ABC-DLBCL patients.


Subject(s)
HSP110 Heat-Shock Proteins , Lymphoma, Large B-Cell, Diffuse , Receptors, Antigen, B-Cell , Signal Transduction , Syk Kinase , Xenograft Model Antitumor Assays , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Syk Kinase/antagonists & inhibitors , Syk Kinase/metabolism , Humans , Phosphorylation/drug effects , Animals , Mice , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , HSP110 Heat-Shock Proteins/metabolism , Pyrimidines/pharmacology , Cell Line, Tumor , Tumor Cells, Cultured , Mice, SCID , Quinazolines
SELECTION OF CITATIONS
SEARCH DETAIL