Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.315
Filter
1.
Rapid Commun Mass Spectrom ; 38(19): e9878, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39117991

ABSTRACT

RATIONALE: Natural variations in the abundance of the stable isotopes of nitrogen (δ15N) and carbon (δ13C) offer valuable insights into metabolic fluxes. In the wake of strong interest in cancer metabolism, recent research has revealed δ15N and δ13C variations in cancerous compared to non-cancerous tissues and cell lines. However, our understanding of natural isotopic variations in cultured mammalian cells, particularly in relation to metabolism, remains limited. This study aims to start addressing this gap using metabolic modulations in cells cultured under controlled conditions. METHODS: Prostate cancer cells (PC3) were cultured in different conditions and their δ15N and δ13C were measured using isotope ratio mass spectrometry. Isotopic variations during successive cell culture passages were assessed and two widely used cell culture media (RPMI and DMEM) were compared. Metabolism was modulated through glutamine deprivation and hypoxia. RESULTS: Successive cell culture passages generally resulted in reproducible δ15N and δ13C values. The impact of culture medium composition on δ15N and δ13C of the cells highlights the importance of maintaining a consistent medium composition across conditions whenever possible. Glutamine deprivation and hypoxia induced a lower δ13C in bulk cell samples, with only the former affecting δ15N. Gaps between theory and experiments were bridged and the lessons learned throughout the process are provided. CONCLUSIONS: Exposing cultured cancer cells to hypoxia allowed us to further investigate the relation between metabolic modulations and natural isotopic variations, while mitigating the confounding impact of changing culture medium composition. This study highlights the potential of natural δ13C variations for studying substrate fluxes and nutrient allocation in reproducible culture conditions. Considering cell yield and culture medium composition is pivotal to the success of this approach.


Subject(s)
Carbon Isotopes , Culture Media , Mass Spectrometry , Nitrogen Isotopes , Humans , Carbon Isotopes/analysis , Carbon Isotopes/metabolism , Nitrogen Isotopes/analysis , Nitrogen Isotopes/metabolism , Mass Spectrometry/methods , Culture Media/chemistry , Culture Media/metabolism , Glutamine/metabolism , Glutamine/analysis , Prostatic Neoplasms/metabolism , Male , PC-3 Cells , Cell Line, Tumor , Cell Culture Techniques/methods
2.
Biomolecules ; 14(8)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39199385

ABSTRACT

Clinically, prostate cancer is infamous for its histological and molecular heterogeneity, which causes great challenges to pinpoint therapy and pharmaceutical development. To overcome these difficulties, researchers are focusing on modulating tumor microenvironment and immune responses in addition to genetic alteration and epigenetic regulation. Here, we aimed to identify potential biomarkers or modulators of prostate cancer by investigating genes specifically altered in prostate cancer cells treated with established anti-cancer agents. Glycerol kinase 1 (GK1) is phosphotransferase encoded on the X chromosome, is associated with the synthesis of triglycerides and glycerophospholipids, and has been mainly studied for X-linked metabolic disorder GK deficiency (GKD). Interestingly, our DNA microarray analysis showed that several anti-cancer agents highly induced the expression of GK1, especially GK1a and GK1b isoforms, in human prostate cancer PC-3 cells. To elucidate the relationship between GK1 and cancer cell death, a human GK1b-specific expression vector was constructed and transfected into the PC-3 cells. Surprisingly, GK1b overexpression dramatically reduced cell viability and significantly accelerated apoptotic cell death. These findings suggest that GK1b may serve as a promising modulator and biomarker of cell death in prostate cancer, offering potential avenues for therapeutic intervention.


Subject(s)
Glycerol Kinase , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Glycerol Kinase/metabolism , Glycerol Kinase/genetics , PC-3 Cells , Gene Expression Regulation, Neoplastic , Apoptosis/genetics , Cell Survival , Antineoplastic Agents/pharmacology , Cell Line, Tumor
3.
Sci Rep ; 14(1): 16047, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38992105

ABSTRACT

ß-glucans are polysaccharides found in the cell walls of various fungi, bacteria and cereals. ß-glucan have been found to show various kinds of anti-inflammatory, antimicrobial, antidiabetic antioxidant and anticancerous activities. In the present study, we have isolated ß-glucan from the baker's yeast Saccharomyces cerevisiae and white button mushroom Agaricus bisporus and tested their antioxidant potential and anticancerous activity against prostate cancer cell line PC3. Particles were characterized with zeta sizer and further with FTIR that confirmed that the isolated particles are ß-glucan and alginate sealing made slow and sustained release of the Quercetin from the ß-glucan particles. Morphological analysis of the hollow and Quercetin loaded ß-glucan was performed with the SEM analysis and stability was analyzed with TGA and DSC analysis that showed the higher stability of the alginate sealed particles. Assessments of the antioxidant potential showed that Quercetin loaded particles were having higher antioxidant activity than hollow ß-glucan particles. Cell viability of the PC3 cells was examined with MTT assay and it was found that Quercetin loaded alginate sealed Agaricus bisporus derived ß-glucan particles were having lowest IC50. Further ROS generation was found to increase in a dose dependent manner. Apoptosis detection was carried out with Propidium iodide and AO/EtBr staining dye which showed significant death in the cells treated with higher concentration of the particles. Study showed that particles derived from both of the sources were having efficient anticancer activity and showing a dose dependent increase in cell death in PC3 cells upon treatment.


Subject(s)
Agaricus , Antineoplastic Agents , Antioxidants , Quercetin , Saccharomyces cerevisiae , beta-Glucans , Quercetin/pharmacology , Quercetin/chemistry , beta-Glucans/pharmacology , beta-Glucans/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Agaricus/chemistry , Saccharomyces cerevisiae/drug effects , Cell Survival/drug effects , PC-3 Cells , Cell Line, Tumor , Reactive Oxygen Species/metabolism
4.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062984

ABSTRACT

For many years, it has been speculated that elevated testosterone levels may be critically involved in the genesis and proliferation of prostate cancer. METHODS: The effect of testosterone on the metabolic activity of hormone-independent [PC-3] and hormone-dependent [LNCAP] cancer cells was investigated in vitro. Additionally, the impact of testosterone nanoemulsion [nanocare®] on cell viability was accessed by flow cytometry. RESULTS: Despite the dependency of the normal prostate and of most prostatic cancers upon androgens, the obtained results indicate that, contrary to prevailing opinion, the supplementation of testosterone with higher doses in nanoemulsion was able to lower the metabolic activity and viability of prostate cancer cells. CONCLUSIONS: We conclude that the growth of hormone-independent and hormone-dependent prostate cancer cells was reduced by the exposure of a nanoemulsion of bioidentical testostosterone in vitro. To the best of our knowledge, this is the first time that the potential effect of a testosterone nanoemulsion on the metabolic activity of prostate cancer cells has been shown. Such tests suggest that the growth of hormone-independent and hormone-dependent prostate cancer cells was reduced by the administration of bioidentical testostosterone, and this might be an interesting strategy for prostate cancer treatment in diagnosed patients.


Subject(s)
Cell Survival , Emulsions , Prostatic Neoplasms , Testosterone , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Testosterone/pharmacology , Cell Survival/drug effects , Cell Line, Tumor , Nanoparticles/chemistry , Cell Proliferation/drug effects , PC-3 Cells , Androgens/pharmacology
5.
Biomed Pharmacother ; 177: 117125, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002444

ABSTRACT

Active targeting to cancer involves exploiting specific interactions between receptors on the surface of cancer cells and targeting moieties conjugated to the surface of vectors such that site-specific delivery is achieved. Prostate specific membrane antigen (PSMA) has proved to be an excellent target for active targeting to prostate cancer. We report the synthesis and use of a PSMA-specific ligand (Glu-NH-CO-NH-Lys) for the site-specific delivery of brusatol- and docetaxel-loaded poly(lactide-co-glycolide) (PLGA) nanoparticles to prostate cancer. The PSMA targeting ligand covalently linked to PLGA-PEG3400 was blended with methoxyPEG-PLGA to prepare brusatol- and docetaxel-loaded nanoparticles with different surface densities of the targeting ligand. Flow cytometry was used to evaluate the impact of different surface densities of the PSMA targeting ligand in LNCaP prostate cancer cells at 15 min and 2 h. Cytotoxicity evaluations of the targeted nanoparticles reveal differences based on PSMA expression in PC-3 and LNCaP cells. In addition, levels of reactive oxygen species (ROS) were measured using the fluorescent indicator, H2DCFDA, by flow cytometry. PSMA-targeted nanoparticles loaded with docetaxel and brusatol showed increased ROS generation in LNCaP cells compared to PC-3 at different time points. Furthermore, the targeted nanoparticles were evaluated in male athymic BALB/c mice implanted with PSMA-producing LNCaP cell tumors. Evaluation of the percent relative tumor volume show that brusatol-containing nanoparticles show great promise in inhibiting tumor growth. Our data also suggest that the dual drug-loaded targeted nanoparticle platform improves the efficacy of docetaxel in male athymic BALB/c mice implanted with PSMA-producing LNCaP cell tumors.


Subject(s)
Antigens, Surface , Docetaxel , Glutamate Carboxypeptidase II , Nanoparticles , Prostatic Neoplasms , Male , Docetaxel/pharmacology , Docetaxel/administration & dosage , Animals , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism , Cell Line, Tumor , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , PC-3 Cells , Mice , Xenograft Model Antitumor Assays , Zebrafish , Mice, Nude , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Mice, Inbred BALB C , Nanoparticle Drug Delivery System/chemistry
6.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000112

ABSTRACT

Androgen-receptor-negative, androgen-independent (ARneg-AI) prostate cancer aggressively proliferates and metastasizes, which makes treatment difficult. Hence, it is necessary to continue exploring cancer-associated markers, such as oncofetal Receptor Tyrosine Kinase like Orphan Receptor 1 (ROR1), which may serve as a form of targeted prostate cancer therapy. In this study, we identify that Penta-O-galloyl-ß-D-glucose (PGG), a plant-derived gallotannin small molecule inhibitor, modulates ROR1-mediated oncogenic signaling and mitigates prostate cancer phenotypes. Results indicate that ROR1 protein levels were elevated in the highly aggressive ARneg-AI PC3 cancer cell line. PGG was selectively cytotoxic to PC3 cells and induced apoptosis of PC3 (IC50 of 31.64 µM) in comparison to normal prostate epithelial RWPE-1 cells (IC50 of 74.55 µM). PGG was found to suppress ROR1 and downstream oncogenic pathways in PC3 cells. These molecular phenomena were corroborated by reduced migration, invasion, and cell cycle progression of PC3 cells. PGG minimally and moderately affected RWPE-1 and ARneg-AI DU145, respectively, which may be due to these cells having lower levels of ROR1 expression in comparison to PC3 cells. Additionally, PGG acted synergistically with the standard chemotherapeutic agent docetaxel to lower the IC50 of both compounds about five-fold (combination index = 0.402) in PC3 cells. These results suggest that ROR1 is a key oncogenic driver and a promising target in aggressive prostate cancers that lack a targetable androgen receptor. Furthermore, PGG may be a selective and potent anti-cancer agent capable of treating ROR1-expressing prostate cancers.


Subject(s)
Cell Proliferation , Glycogen Synthase Kinase 3 beta , Hydrolyzable Tannins , Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Receptor Tyrosine Kinase-like Orphan Receptors , Signal Transduction , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Hydrolyzable Tannins/pharmacology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , PC-3 Cells , Gene Expression Regulation, Neoplastic/drug effects , Docetaxel/pharmacology
7.
Molecules ; 29(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38999000

ABSTRACT

In this study, a new series of cis and trans 5-substituted-3-(dibenzyloxyphosphoryl)isoxazolidines 16a-g were synthesized by the 1,3-dipolar cycloaddition reaction of N-benzyl-C-(dibenzyloxyphosphoryl)nitrone and selected N1-allyl-N3-benzylquinazoline-2,4-diones. All the obtained trans-isoxazolidines 16a-g and the samples enriched in respective cis-isomers were evaluated for anticancer activity against three tumor cell lines. All the tested compounds exhibited high activity against the prostate cancer cell line (PC-3). Isoxazolidines trans-16a and trans-16b and diastereoisomeric mixtures of isoxazolidines enriched in cis-isomer using HPLC, namely cis-16a/trans-16a (97:3) and cis-16b/trans-16b (90:10), showed the highest antiproliferative properties towards the PC-3 cell line (IC50 = 9.84 ± 3.69-12.67 ± 3.45 µM). For the most active compounds, induction apoptosis tests and an evaluation of toxicity were conducted. Isoxazolidine trans-16b showed the highest induction of apoptosis. Moreover, the most active compounds turned out safe in vitro as none affected the cell viability in the HEK293, HepG2, and HSF cellular models at all the tested concentrations. The results indicated isoxazolidine trans-16b as a promising new lead structure in the search for effective anticancer drugs.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Structure-Activity Relationship , Isoxazoles/chemistry , Isoxazoles/pharmacology , PC-3 Cells , Drug Screening Assays, Antitumor , Quinazolinones/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemical synthesis , Molecular Structure , Cell Survival/drug effects , Apoptosis/drug effects
8.
J Nanobiotechnology ; 22(1): 391, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965509

ABSTRACT

BACKGROUND: Prostate cancer (PCa) has a high incidence in men worldwide, and almost all PCa patients progress to the androgen-independent stage which lacks effective treatment measures. PTENP1, a long non-coding RNA, has been shown to suppress tumor growth through the rescuing of PTEN expression via a competitive endogenous RNA (ceRNA) mechanism. However, PTENP1 was limited to be applied in the treatment of PCa for the reason of rapid enzymatic degradation, poor intracellular uptake, and excessively long base sequence to be synthesized. Considering the unique advantages of artificial nanomaterials in drug loading and transport, black phosphorus (BP) nanosheet was employed as a gene-drug carrier in this study. RESULTS: The sequence of PTENP1 was adopted as a template which was randomly divided into four segments with a length of about 1000 nucleotide bases to synthesize four different RNA fragments as gene drugs, and loaded onto polyethyleneimine (PEI)-modified BP nanosheets to construct BP-PEI@RNA delivery platforms. The RNAs could be effectively delivered into PC3 cells by BP-PEI nanosheets and elevating PTEN expression by competitive binding microRNAs (miRNAs) which target PTEN mRNA, ultimately exerting anti-tumor effects. CONCLUSIONS: Therefore, this study demonstrated that BP-PEI@RNAs is a promising gene therapeutic platform for PCa treatment.


Subject(s)
Nanostructures , PTEN Phosphohydrolase , Phosphorus , Prostatic Neoplasms , Male , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Phosphorus/chemistry , Nanostructures/chemistry , MicroRNAs/genetics , Cell Line, Tumor , PC-3 Cells , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Polyethyleneimine/chemistry , Animals , Gene Transfer Techniques , Gene Expression Regulation, Neoplastic/drug effects , RNA, Competitive Endogenous
9.
J Med Virol ; 96(7): e29748, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38975633

ABSTRACT

Prostate cancer is a prevalent carcinoma among males, and conventional treatment options are often limited. Cytotoxic chemotherapy, despite its drawbacks, remains a mainstay. We propose a targeted co-delivery approach using nanoscale delivery units for Oncolytic measles virus (OMV) and vincristine (VC) to enhance treatment efficacy. The HA-coated OMV + VC-loaded TCs nanoformulation is designed for targeted oncolytic activity in prostate cancer. The CD44 expression analysis in prostate cancer cell lines indicates a significantly high expression in PC3 cells. The optimization of nanoformulations using Design of Expert (DOE) is performed, and the preparation and characterization of HA-coated OMV + VC-loaded TCs nanoformulations are detailed showing average particle size 397.2 ± 0.01 nm and polydispersity index 0.122 with zeta potential 19.7 + 0.01 mV. Results demonstrate successful encapsulation efficiency with 2.4 × 106 TCID50/Ml and sustained release of OMV and VC from the nanoformulation for up to 72 h. In vitro, assays reveal potent anticancer activity at 10 ± 0.71% cell viability in PC3 cells compared to 73 ± 0.66% in HPrEC and significant morphological changes at 90 µg/ml in dose and time-dependent manner. The co-formulation showed positive cell death 49.5 ± 0.02% at 50 µg PI/ml in PBS and 54.3% cell cycle arrest at the G2/M phase, 8.1% G0/G1 and 5.7% at S phase, with significant mitochondrial membrane potential (MMP) at 50 µg/ml, as assessed by flow cytometry (FACS). The surface-integrating ligand approach enhances the targeted delivery of the oncolytic virus and chemotherapeutic drug, presenting a potential alternative for prostate cancer treatment and suggested that co-administering VC and OMV in a nanoformulation could improve therapeutic outcomes while reducing chemotherapeutic drug doses.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Prostatic Neoplasms , Vincristine , Humans , Male , Prostatic Neoplasms/therapy , Prostatic Neoplasms/drug therapy , Vincristine/pharmacology , Vincristine/administration & dosage , Oncolytic Virotherapy/methods , Cell Line, Tumor , Measles virus/drug effects , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Nanoparticles/chemistry , PC-3 Cells
10.
Toxicol Appl Pharmacol ; 490: 117021, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971382

ABSTRACT

Prostate cancer is a common type of cancer in men with high incidence and mortality. Our aim was to investigate the effects of oxalipalladium (ox-Pd) on metastatic human prostate cancer PC3 cells and compare them with the effects of oxaliplatin (ox-Pt) (as an approved cancer drug). We synthesized ox-Pd through a new chemical method and used FT-IR, 1H NMR, 13C NMR, and MS analyzes to characterize it. The effects of ox-Pd on PC3 cells viability, apoptosis, cell cycle, migration, and gene expression were examined. Inhibition of topoisomerase IIα activity was investigated by pHOT1 plasmid relaxation and kDNA decatenation assays. Chemical tests showed ox-Pd with the correct composition and structure. For the first time, the exact fragmentation pathway of ox-Pd and its difference with ox-Pt was obtained by MS analysis. Ox-Pd significantly decreased PC3 cell viability with less/no toxicity effect on MHFB-1 normal skin fibroblasts. Wound scratch assay confirmed the strong anti-migratory activity of ox-Pd. According to flow cytometry analysis, this drug increased the number of PC3 cells in late apoptosis and decreased DNA replication and mitosis. Furthermore, pHOT1 plasmid relaxation and kDNA decatenation assays showed that ox-Pd strongly inhibited the catalytic activity of topoisomerase IIα. The expression of topoisomerase IIα, Bcl-2, P21, and survivin was decreased while the expression of Bax and p53 was increased under ox-Pd treatment. We provide the first evidence that ox-Pd exhibits more selective anticancer effects on PC3 cells compared to ox-Pt. Taken together, these data strongly suggest a therapeutic window for ox-Pd in cancer.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Survival , Prostatic Neoplasms , Humans , Male , Antineoplastic Agents/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , PC-3 Cells , Apoptosis/drug effects , Cell Survival/drug effects , Cell Movement/drug effects , Palladium/pharmacology , Palladium/chemistry , Oxaliplatin/pharmacology , Cell Cycle/drug effects
11.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891892

ABSTRACT

Recently, a compound derived from recent scientific advances named 34 has emerged as the focus of this research, the aim of which is to explore its potential impact on solid tumor cell lines. Using a combination of bioinformatics and biological assays, this study conducted an in-depth investigation of the effects of 34. The results of this study have substantial implications for cancer research and treatment. 34 has shown remarkable efficacy in inhibiting the growth of several cancer cell lines, including those representing prostate carcinoma (PC3) and cervical carcinoma (HeLa). The high sensitivity of these cells, indicated by low IC50 values, underscores its potential as a promising chemotherapeutic agent. In addition, 34 has revealed the ability to induce cell cycle arrest, particularly in the G2/M phase, a phenomenon with critical implications for tumor initiation and growth. By interfering with DNA replication in cancer cells, 34 has shown the capacity to trigger cell death, offering a new avenue for cancer treatment. In addition, computational analyses have identified key genes affected by 34 treatment, suggesting potential therapeutic targets. These genes are involved in critical biological processes, including cell cycle regulation, DNA replication and microtubule dynamics, all of which are central to cancer development and progression. In conclusion, this study highlights the different mechanisms of 34 that inhibit cancer cell growth and alter the cell cycle. These promising results suggest the potential for more effective and less toxic anticancer therapies. Further in vivo validation and exploration of combination therapies are critical to improve cancer treatment outcomes.


Subject(s)
Acrylonitrile , Antineoplastic Agents , Microtubules , Humans , Microtubules/drug effects , Microtubules/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Acrylonitrile/analogs & derivatives , Acrylonitrile/pharmacology , Acrylonitrile/therapeutic use , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , HeLa Cells , Apoptosis/drug effects , Triazoles/pharmacology , Cell Cycle Checkpoints/drug effects , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , PC-3 Cells
12.
Mol Pharm ; 21(8): 4128-4146, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38920398

ABSTRACT

Prostate cancer is a prevalently detected malignancy with a dismal prognosis. Luteinizing-hormone-releasing-hormone (LHRH) receptors are overexpressed in such cancer cells, to which the LHRH-decapeptide can specifically bind. A lipid-polyethylene glycol-conjugated new LHRH-decapeptide analogue (D-P-HLH) was synthesized and characterized. D-P-HLH-coated and anticancer drug doxorubicin (DX)-loaded solid lipid nanoparticles (F-DX-SLN) were formulated by the cold homogenization technique and characterized by Fourier transform infrared spectroscopy, X-ray diffraction, X-ray photoelectron spectroscopy, differential scanning calorimetry, dynamic light scattering, electron microscopy, entrapment efficiency, and drug-release profile studies. F-DX-SLN allows site-specific DX delivery by reducing the side effects of chemotherapy. Cancer cells could precisely take up F-DX-SLN by targeting specific receptors, boosting the cytotoxicity at the tumor site. The efficacy of F-DX-SLN on PC3/SKBR3 cells by the MTT assay revealed that F-DX-SLN was more cytotoxic than DX and/or DX-SLN. Flow cytometry and confocal microscopic studies further support F-DX-SLNs' increased intracellular absorption capability in targeting LHRH overexpressed cancer cells. F-DX-SLN ensured high apoptotic potential, noticeably larger mitochondrial transmembrane depolarization action, as well as the activation of caspases, a longer half-life, and greater plasma concentration. F-DX-SLN/DX-SLN was radiolabeled with technetium-99m; scintigraphic imaging studies established its tumor selectivity in PC3 tumor-bearing nude mice. The efficacy of the formulations in cancer treatment, in vivo therapeutic efficacy tests, and histopathological studies were also conducted. Results clearly indicate that F-DX-SLN exhibits sustained and superior targeted administration of anticancer drugs, thus opening up the possibility of a drug delivery system with precise control and targeting effects. F-DX-SLN could also provide a nanotheranostic approach with improved efficacy for prostate cancer therapy.


Subject(s)
Doxorubicin , Gonadotropin-Releasing Hormone , Lipids , Nanoparticles , Prostatic Neoplasms , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Male , Animals , Gonadotropin-Releasing Hormone/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/chemistry , Nanoparticles/chemistry , Mice , Cell Line, Tumor , Lipids/chemistry , Mice, Nude , Drug Carriers/chemistry , Polyethylene Glycols/chemistry , Drug Liberation , PC-3 Cells , Receptors, LHRH/metabolism , Drug Delivery Systems/methods , Apoptosis/drug effects
13.
Planta Med ; 90(10): 774-784, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942031

ABSTRACT

Hyaluronic acid is composed of repeating sugar units, glucuronic acid and N-acetylglucosamine, which are often associated with increased tumor progression. Urtica dioica agglutinin is a potential component that exhibits a high affinity for binding to N-acetylglucosamine. This study aimed to investigate U. dioica Agglutinin's potential to inhibit the proliferation and migration of prostate cancer cells with high expression of hyaluronic acid through molecular docking and in vitro studies. The expression of hyaluronan synthase genes in prostate tissue and cell lines was checked by an in silico study, and the interaction between hyaluronic acid with both CD44 transmembrane glycoprotein and U. dioica agglutinin was analyzed through molecular docking. U. dioica Agglutinin's effect on cell viability (neutral red uptake assay), migration (scratch wound healing assays), and both CD44 and Nanog expression (quantitative real-time polymerase chain reaction) were assessed in vitro. The results showed that in prostate cancer cell lines, the PC3 cell line has the highest expression of hyaluronan synthase genes. U. dioica agglutinin exhibits an interaction of six specific residues on CD44 compared to hyaluronic acid's singular residue. While U. dioica agglutinin alone effectively reduced cell viability and wound closer (≥ 150 µg/mL), combining it with hyaluronic acid significantly shifted the effective concentration to a higher dose (≥ 350 µg/mL). These results, together with low Nanog and high CD44 gene expression, suggest that U. dioica agglutinin may impair the CD44-HA pathway in PC3 cells. This possibility is supported by U. dioica Agglutinin's ability to compete with hyaluronic acid for binding to CD44. Based on this, U. dioica agglutinin as a plant lectin shows promise in inhibiting cancer proliferation and migration by targeting its dependence on hyaluronic acid.


Subject(s)
Cell Movement , Cell Proliferation , Hyaluronan Receptors , Hyaluronic Acid , Prostatic Neoplasms , Urtica dioica , Humans , Hyaluronic Acid/pharmacology , Male , Prostatic Neoplasms/drug therapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Urtica dioica/chemistry , Cell Line, Tumor , Hyaluronan Receptors/metabolism , Molecular Docking Simulation , Cell Survival/drug effects , Agglutinins/pharmacology , Hyaluronan Synthases/metabolism , PC-3 Cells
14.
Int J Mol Sci ; 25(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892219

ABSTRACT

Anoctamin1 (ANO1), a calcium-activated chloride channel, is overexpressed in a variety of cancer cells, including prostate cancer, and is involved in cancer cell proliferation, migration, and invasion. Inhibition of ANO1 in these cancer cells exhibits anticancer effects. In this study, we conducted a screening to identify novel ANO1 inhibitors with anticancer effects using PC-3 human prostate carcinoma cells. Screening of 2978 approved and investigational drugs revealed that hemin is a novel ANO1 inhibitor with an IC50 value of 0.45 µM. Notably, hemin had no significant effect on intracellular calcium signaling and cystic fibrosis transmembrane conductance regulator (CFTR), a cyclic AMP (cAMP)-regulated chloride channel, and it showed a weak inhibitory effect on ANO2 at 3 µM, a concentration that completely inhibits ANO1. Interestingly, hemin also significantly decreased ANO1 protein levels and strongly inhibited the cell proliferation and migration of PC-3 cells in an ANO1-dependent manner. Furthermore, it strongly induced caspase-3 activation, PARP degradation, and apoptosis in PC-3 cells. These findings suggest that hemin possesses anticancer properties via ANO1 inhibition and could be considered for development as a novel treatment for prostate cancer.


Subject(s)
Anoctamin-1 , Antineoplastic Agents , Hemin , Neoplasm Proteins , Prostatic Neoplasms , Humans , Male , Anoctamin-1/metabolism , Anoctamin-1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Hemin/pharmacology , Neoplasm Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
15.
Molecules ; 29(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38930989

ABSTRACT

The crystal structures of two newly synthesized nitrilotriacetate oxidovanadium(IV) salts, namely [QH][VO(nta)(H2O)](H2O)2 (I) and [(acr)H][VO(nta)(H2O)](H2O)2 (II), were determined. Additionally, the cytotoxic effects of four N-heterocyclic nitrilotriacetate oxidovanadium(IV) salts-1,10-phenanthrolinium, [(phen)H][VO(nta)(H2O)](H2O)0.5 (III), 2,2'-bipyridinium [(bpy)H][VO(nta)(H2O)](H2O) (IV), and two newly synthesized compounds (I) and (II)-were evaluated against prostate cancer (PC3) and breast cancer (MCF-7) cells. All the compounds exhibited strong cytotoxic effects on cancer cells and normal cells (HaCaT human keratinocytes). The structure-activity relationship analysis revealed that the number and arrangement of conjugated aromatic rings in the counterion had an impact on the antitumor effect. The compound (III), the 1,10-phenanthrolinium analogue, exhibited the greatest activity, whereas the acridinium salt (II), with a different arrangement of three conjugated aromatic rings, showed the lowest toxicity. The increased concentrations of the compounds resulted in alterations to the cell cycle distribution with different effects in MCF-7 and PC3 cells. In MCF-7 cells, compounds I and II were observed to block the G2/M phase, while compounds III and IV were found to arrest the cell cycle in the G0/G1 phase. In PC3 cells, all compounds increased the rates of cells in the G0/G1 phase.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Male , Female , MCF-7 Cells , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Nitrilotriacetic Acid/chemistry , Nitrilotriacetic Acid/analogs & derivatives , Structure-Activity Relationship , Cell Line, Tumor , Cell Proliferation/drug effects , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Vanadium/chemistry , Vanadium/pharmacology , PC-3 Cells , Cell Cycle/drug effects , Molecular Structure , Salts/chemistry , Salts/pharmacology , Cell Survival/drug effects , Apoptosis/drug effects
16.
Nutrients ; 16(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38931175

ABSTRACT

Cancer therapy, from malignant tumor inhibition to cellular eradication treatment, remains a challenge, especially regarding reduced side effects and low energy consumption during treatment. Hence, phytochemicals as cytotoxic sensitizers or photosensitizers deserve special attention. The dark and photo-response of Yemenite 'Etrog' leaf extracts applied to prostate PC3 cancer cells is reported here. An XTT cell viability assay along with light microscope observations revealed pronounced cytotoxic activity of the extract for long exposure times of 72 h upon concentrations of 175 µg/mL and 87.5 µg/mL, while phototoxic effect was obtained even at low concentration of 10.93 µg/mL and a short introduction period of 1.5 h. For the longest time incubation of 72 h and for the highest extract concentration of 175 µg/mL, relative cell survival decreased by up to 60% (below the IC50). In combined phyto-photodynamic therapy, a reduction of 63% compared to unirradiated controls was obtained. The concentration of extract in cells versus the accumulation time was inversely related to fluorescence emission intensity readings. Extracellular ROS production was also shown. Based on an ATR-FTIR analysis of the powdered leaves and their liquid ethanolic extract, biochemical fingerprints of both polar and non-polar phyto-constituents were identified, thereby suggesting their implementation as phyto-medicine and phyto-photomedicine.


Subject(s)
Cell Survival , Photochemotherapy , Plant Extracts , Plant Leaves , Prostatic Neoplasms , Humans , Male , Plant Extracts/pharmacology , Photochemotherapy/methods , Prostatic Neoplasms/drug therapy , Plant Leaves/chemistry , Cell Survival/drug effects , Photosensitizing Agents/pharmacology , PC-3 Cells , Reactive Oxygen Species/metabolism , Yemen , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/pharmacology
17.
Spectrochim Acta A Mol Biomol Spectrosc ; 320: 124624, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-38878725

ABSTRACT

Biscarbazole derivative probe (6) (Z)-2-(3-(((9-heptyl-9H-carbazol-3-yl)methylene)amino)-9H-carbazol-9-yl)ethan-1-ol containing an imine group, which is a sensitive and selective fluorescence chemosensor, was designed and synthesized for the effective evaluation of Cu2+ metal ion levels. The synthesized compounds were characterized using 1H NMR, 13C NMR, FT-IR, and MALDI-TOF MS (for compound 6) spectroscopic data. The interaction model between probe 6 and Cu2+ was determined by combining fluorescence methods, 1H NMR titration, Job's plot, and theoretical calculations. For probe 6, the fluorogenic recognition of Cu2+ was investigated by fluorescence spectroscopy, and the optical changes caused by Cu2+ ions were carried out in ACN/H2O (50:50) solution at pH 7.0. Fluorescence probe 6 was found to "turn-off" its fluorescence in the presence of paramagnetic Cu2+ ions. Probe 6 was determined to have a rapid response within 40s and showed a fluorescence response to Cu2+ with a low detection limit of 0.16 µM. Additionally, in vitro anticancer activity and cell imaging studies of probe 6 against the prostate cell line (PC-3) were performed.


Subject(s)
Antineoplastic Agents , Carbazoles , Copper , Fluorescent Dyes , Spectrometry, Fluorescence , Copper/chemistry , Copper/analysis , Humans , Carbazoles/chemistry , Carbazoles/chemical synthesis , Carbazoles/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacology , Fluorometry/methods , Cell Line, Tumor , PC-3 Cells
18.
Biomed Pharmacother ; 176: 116887, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852511

ABSTRACT

BACKGROUND: The metastasis of tumors into bone tissue typically leads to intractable pain that is both very disabling and particularly difficult to manage. We investigated here whether riluzole could have beneficial effects for the treatment of prostate cancer-induced bone pain and how it could influence the development of bone metastasis. METHODS: We used a bone pain model induced by intratibial injection of human PC3 prostate cancer cells into male SCID mice treated or not with riluzole administered in drinking water. We also used riluzole in vitro to assess its possible effect on PC3 cell viability and functionality, using patch-clamp. RESULTS: Riluzole had a significant preventive effect on both evoked and spontaneous pain involving the TREK-1 potassium channel. Riluzole did not interfere with PC3-induced bone loss or bone remodeling in vivo. It also significantly decreased PC3 cell viability in vitro. The antiproliferative effect of riluzole is correlated with a TREK-1-dependent membrane hyperpolarization in these cells. CONCLUSION: The present data suggest that riluzole could be very useful to manage evoked and spontaneous hypersensitivity in cancer-induced bone pain and has no significant adverse effect on cancer progression.


Subject(s)
Analgesics , Bone Neoplasms , Cancer Pain , Cell Proliferation , Mice, SCID , Potassium Channels, Tandem Pore Domain , Riluzole , Riluzole/pharmacology , Animals , Potassium Channels, Tandem Pore Domain/metabolism , Male , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Bone Neoplasms/pathology , Bone Neoplasms/complications , Humans , Cancer Pain/drug therapy , Cancer Pain/metabolism , Analgesics/pharmacology , Cell Proliferation/drug effects , PC-3 Cells , Mice , Cell Survival/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Cell Line, Tumor
19.
Sci Signal ; 17(840): eadc9142, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861615

ABSTRACT

Neuroendocrine prostate cancer (PCa) (NEPC), an aggressive subtype that is associated with poor prognosis, may arise after androgen deprivation therapy (ADT). We investigated the molecular mechanisms by which ADT induces neuroendocrine differentiation in advanced PCa. We found that transmembrane protein 1 (MCTP1), which has putative Ca2+ sensing function and multiple Ca2+-binding C2 domains, was abundant in samples from patients with advanced PCa. MCTP1 was associated with the expression of the EMT-associated transcription factors ZBTB46, FOXA2, and HIF1A. The increased abundance of MCTP1 promoted PC3 prostate cancer cell migration and neuroendocrine differentiation and was associated with SNAI1-dependent EMT in C4-2 PCa cells after ADT. ZBTB46 interacted with FOXA2 and HIF1A and increased the abundance of MCTP1 in a hypoxia-dependent manner. MCTP1 stimulated Ca2+ signaling and AKT activation to promote EMT and neuroendocrine differentiation by increasing the SNAI1-dependent expression of EMT and neuroendocrine markers, effects that were blocked by knockdown of MCTP1. These data suggest an oncogenic role for MCTP1 in the maintenance of a rare and aggressive prostate cancer subtype through its response to Ca2+ and suggest its potential as a therapeutic target.


Subject(s)
Cell Differentiation , Epithelial-Mesenchymal Transition , Prostatic Neoplasms , Animals , Humans , Male , Mice , Androgens/metabolism , Androgens/pharmacology , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
20.
Int J Mol Sci ; 25(10)2024 May 18.
Article in English | MEDLINE | ID: mdl-38791557

ABSTRACT

A promising de novo approach for the treatment of Castration-resistant prostate cancer (CRPC) exploits cell-mediated enzyme prodrug therapy comprising cytosine deaminase (CD) and fluorouracil (5-FC). The aim of this study was to determine the potential of bacterial CD-overexpressing hTERT-immortalized human adipose stem cells (hTERT-ADSC.CD) to suppress CRPC. A lentiviral vector encoding a bacterial CD gene was used to transfect and to generate the hTERT-ADSC.CD line. The ability of the cells to migrate selectively towards malignant cells was investigated in vitro. PC3 and hTERT-ADSC.CD cells were co-cultured. hTERT-ADSC.CD and 1 × 106 PC3 cells were administered to nude mice via intracardiac and subcutaneous injections, respectively, and 5-FC was given for 14 days. hTERT-ADSC.CD were successfully engineered. Enhanced in vitro hTERT-ADSC.CD cytotoxicity and suicide effect were evident following administration of 5 µM 5-FC. hTERT-ADSC.CD, together with 5-FC, augmented the numbers of PC3 cells undergoing apoptosis. In comparison to controls administered hTERT-ADSC.CD monotherapy, hTERT-ADSC.CD in combination with 5-FC demonstrated a greater suppressive effect on tumor. In CPRC-bearing mice, tumor suppression was enhanced by the combination of CD-overexpressing ADSC and the prodrug 5-FC. Stem cells exhibiting CD gene expression are a potential novel approach to treatment for CRPC.


Subject(s)
Cytosine Deaminase , Flucytosine , Prostatic Neoplasms, Castration-Resistant , Telomerase , Animals , Humans , Male , Mice , Adipose Tissue/cytology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Flucytosine/pharmacology , Mice, Nude , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/therapy , Stem Cells , Telomerase/genetics , Telomerase/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL