Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86.044
Filter
1.
Mol Biol Rep ; 51(1): 1002, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39305332

ABSTRACT

BACKGROUND: Recent 23Na-MRI reports show higher salt deposition in malignant breast tissue than in surrounding normal tissue. The effect of high salt on cancer progression remains controversial. Here, we investigated the direct effect of high salt on breast cancer progression in vitro. METHODS: Here, the impact of high salt on apoptosis, proliferation, cell cycle, adhesion, and migration of MDA-MB-231 and MCF-7 cells was studied using MTT, scratch, and clonogenic assays, as well as RT-PCR and flow cytometry. Gene expression was analyzed using Real-Time PCR and western blotting. The effect of high salt on global transcriptomics changes in MDA MB-231 cells was studied using RNA-sequencing analysis. RESULTS: Flow cytometry with Annexin V and CFSE revealed that high salt-induced dose-dependent apoptosis and inhibited proliferation. High salt-induced cell cycle arrest at the G1/S phase of the cell cycle. p-MDM2 is known to suppress p53, which plays a crucial role in regulating apoptosis and cell cycle arrest under cellular stress conditions. High salt treatment led to decreased p-MDM2 and increased p53 expression, suggesting that high salt induces apoptosis through p53 stabilization. decreased p-MDM2 and increased p53 expression. High salt also reduced migration and adhesion of cells in a dose-dependent manner suggesting its inhibitory effect on metastatic properties as evident from wound healing assay. RNA sequencing analysis revealed overexpression of tumor suppressor genes and genes associated with anti-tumor activity (PCDHGA11, EIF3CL, RAVER1, TNFSF15, RANBP3L) and under-expression of genes involved in cancer-promoting activity (MT1X, CLDN14, CSF-2). CONCLUSION: Our results unequivocally demonstrate the anti-tumor efficacy of high salt against breast cancer cells, suggesting its potential as a therapeutic strategy in cancer treatment.


Subject(s)
Apoptosis , Breast Neoplasms , Cell Movement , Cell Proliferation , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects , MCF-7 Cells , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Cell Adhesion/drug effects , Cell Cycle/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Sodium Chloride/pharmacology , Cell Cycle Checkpoints/drug effects
2.
Zhongguo Zhong Yao Za Zhi ; 49(15): 4139-4147, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39307746

ABSTRACT

This study aims to explore the effect and mechanism of a mitochondrion-targeted derivative of ergosterol peroxide(Mito-EP) on breast cancer. The methyl thiazolyl tetrazolium(MTT) assay was employed to examine the proliferation of MDA-MB-231 cells treated with different concentrations(0, 0.075, 0.15, 0.3, 0.6, 1.2, and 2.4 µmol·L~(-1)) of Mito-EP. Cells were grouped for treatment with water(blank control), low, medium, and high concentrations(0.15, 0.3, and 0.6 µmol·L~(-1)) of Mito-EP, and ergosterol peroxide(EP)(0.6 µmol·L~(-1)). After the cells were treated for 48 h, flow cytometry was employed to examine the apoptosis rate, reactive oxygen species(ROS) level, mitochondrial membrane potential, and cell cycle distribution, and the apoptosis, ROS, and mitochondrial membrane potential were observed by laser confocal microscopy. A mouse model bearing subcutaneous xenograft tumor was established by injecting 4T1 cell suspension and used to study the inhibitory effect of Mito-EP on breast cancer. Western blot was employed to determine the protein levels of B-cell lymphoma 2(Bcl-2), Bcl-2-associated X protein(Bax), cytochrome C(Cyt C), cleaved caspase-7, and cleaved caspase-9 in cells and the tumor tissue. The results showed that Mito-EP reduced the proliferation rate of MDA-MB-231 cells in a concentration-dependent manner. Compared with the blank control group, EP(0.6 µmol·L~(-1)) caused slight changes in the apoptosis rate, ROS level, and mitochondrial membrane potential. However, Mito-EP increased the apoptosis rate, elevated the ROS level, decreased mitochondrial membrane potential, up-regulated the protein levels of Bax, Cyt C, cleaved caspase-7, and cleaved caspase-9, and down-regulated the protein level of Bcl-2(all P<0.05). Moreover, Mito-EP reduced the tumor volume and weight. In summary, Mito-EP may promote apoptosis in breast cancer cells by activating the mitochondrial apoptosis pathway.


Subject(s)
Apoptosis , Breast Neoplasms , Ergosterol , Mitochondria , Reactive Oxygen Species , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Apoptosis/drug effects , Mice , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Ergosterol/analogs & derivatives , Ergosterol/pharmacology , Cell Proliferation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Nude , Cell Cycle/drug effects
3.
Molecules ; 29(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39274874

ABSTRACT

Activation of the Hedgehog (Hh) signaling pathway is often associated with the progression of various types of cancer. The purpose of study was to search for inhibitors of the Hh signaling pathway among eight compounds belonging to the group of isoxazolyl steroids. The evaluation of the effectiveness of the compounds was based on the analysis of their cytotoxicity, effect on the cell cycle, on the expression of key Hh-signaling-pathway genes (Ptch1, Smo, and Gli1) and putative target genes MMP-2 and MMP-9. Four compounds with the most pronounced cytotoxic effect were identified: compounds 1, 2 (HeLa cells) and 3, 4 (A549 cells). Compounds 1 and 2 significantly reduced the expression of the Ptch1, Smo, Gli1 genes, but had the opposite effect on MMP-2 gene expression: Compound 1 increased it, and compound 2 decreased it. Compounds 3 and 4 did not have a noticeable inhibitory effect on the expression of the Shh pathway receptors, but significantly inhibited MMP-2 and MMP-9 expression. Thus, it was shown that inhibition of the Shh signaling pathway by isoxazolyl steroids can have the opposite effect on MMPs gene expression, which is what should be taken into account in further studies of these compounds as therapeutic agents.


Subject(s)
Gene Expression Regulation, Neoplastic , Hedgehog Proteins , Signal Transduction , Steroids , Humans , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Signal Transduction/drug effects , Steroids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , A549 Cells , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Cell Line, Tumor , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Antineoplastic Agents/pharmacology , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , Smoothened Receptor/antagonists & inhibitors , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Cell Cycle/drug effects
4.
J Cell Biol ; 223(12)2024 Dec 02.
Article in English | MEDLINE | ID: mdl-39316454

ABSTRACT

Subcortical heterotopia is a cortical malformation associated with epilepsy, intellectual disability, and an excessive number of cortical neurons in the white matter. Echinoderm microtubule-associated protein like 1 (EML1) mutations lead to subcortical heterotopia, associated with abnormal radial glia positioning in the cortical wall, prior to malformation onset. This perturbed distribution of proliferative cells is likely to be a critical event for heterotopia formation; however, the underlying mechanisms remain unexplained. This study aimed to decipher the early cellular alterations leading to abnormal radial glia. In a forebrain conditional Eml1 mutant model and human patient cells, primary cilia and centrosomes are altered. Microtubule dynamics and cell cycle kinetics are also abnormal in mouse mutant radial glia. By rescuing microtubule formation in Eml1 mutant embryonic brains, abnormal radial glia delamination and heterotopia volume were significantly reduced. Thus, our new model of subcortical heterotopia reveals the causal link between Eml1's function in microtubule regulation and cell position, both critical for correct cortical development.


Subject(s)
Centrosome , Microtubule-Associated Proteins , Microtubules , Prosencephalon , Animals , Centrosome/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Prosencephalon/metabolism , Prosencephalon/pathology , Prosencephalon/embryology , Microtubules/metabolism , Mice , Cilia/metabolism , Cilia/pathology , Mutation/genetics , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Cell Cycle/genetics
5.
J Agric Food Chem ; 72(38): 20918-20929, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39262347

ABSTRACT

Royal jelly (RJ) is a natural food product with nutritional value and anticancer activity. However, their effects on gastric cancer are unclear. Here, we show that treatment with 5-320 µg/mL of RJ, ethanol extract (RJEE), and protein hydrolyzate (RJPH) decreased the viability of MKN-28 gastric cancer cells, with a half-maximal inhibitory concentration of 123.22 µg/mL for RJEE. RJ, RJEE, and RJPH increase the lactate dehydrogenase release rate and change the morphology of the cells, resulting in cell shrinkage, nucleoplasm condensation, and the formation of apoptotic bodies. RJ and its functional components stagnated the cell cycle in the G0/G1 phase, accompanied by the accumulation of reactive oxygen species, decreased mitochondrial membrane potential, and increased expression levels of p53 and p21 proteins, caspase-3 activation, and apoptosis. Therefore, RJ, RJEE, and RJPH have potential inhibitory effects on the proliferation of gastric cancer cells.


Subject(s)
Apoptosis , Cell Proliferation , Fatty Acids , Stomach Neoplasms , Humans , Stomach Neoplasms/metabolism , Stomach Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Apoptosis/drug effects , Fatty Acids/chemistry , Fatty Acids/pharmacology , Fatty Acids/metabolism , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Caspase 3/metabolism , Caspase 3/genetics , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics
6.
PLoS Biol ; 22(9): e3002791, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39255306

ABSTRACT

Virulence of apicomplexan parasites is based on their ability to divide rapidly to produce significant biomass. The regulation of their cell cycle is therefore key to their pathogenesis. Phosphorylation is a crucial posttranslational modification that regulates many aspects of the eukaryotic cell cycle. The phosphatase PP1 is known to play a major role in the phosphorylation balance in eukaryotes. We explored the role of TgPP1 during the cell cycle of the tachyzoite form of the apicomplexan parasite Toxoplasma gondii. Using a conditional mutant strain, we show that TgPP1 regulates many aspects of the cell cycle including the proper assembly of the daughter cells' inner membrane complex (IMC), the segregation of organelles, and nuclear division. Unexpectedly, depletion of TgPP1 also results in the accumulation of amylopectin, a storage polysaccharide that is usually found in the latent bradyzoite form of the parasite. Using transcriptomics and phospho-proteomics, we show that TgPP1 mainly acts through posttranslational mechanisms by dephosphorylating target proteins including IMC proteins. TgPP1 also dephosphorylates a protein bearing a starch-binding domain. Mutagenesis analysis reveals that the targeted phospho-sites are linked to the ability of the parasite to regulate amylopectin steady-state levels. Therefore, we show that TgPP1 has pleiotropic roles during the tachyzoite cell cycle regulation, but also regulates amylopectin accumulation.


Subject(s)
Amylopectin , Protein Phosphatase 1 , Protozoan Proteins , Toxoplasma , Toxoplasma/metabolism , Toxoplasma/genetics , Toxoplasma/enzymology , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Amylopectin/metabolism , Phosphorylation , Cell Cycle , Animals , Humans
7.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273132

ABSTRACT

Couroupita guianensis, a medicinal plant autochthonal to South America and South India, is widely used in the ethnomedicine of the indigenous peoples of these regions thanks to its alleged antimicrobial, anti-inflammatory, antioxidant and wound-healing properties. The majority of studies have mainly analyzed organic extracts of the Indian plant's flowers and leaves, with limited research on its bark decoction, traditionally used in Amazonian shamanic medicine. In this study, we investigated the anticancer effects of the bark decoction and its main fractions obtained through chromatographic separation, as well as the underlying molecular mechanisms in AGS gastric cancer cells. Viability, cell proliferation, cell cycle, apoptosis and protein expression related to these processes were evaluated. Both the bark decoction and fraction III significantly inhibited cell viability, and the cytotoxic effect was linked to cell cycle blockade and the induction of apoptosis also through an engulfment of the autophagic flux. Increased expression or activation of the key proteins (p53, p21, cdk2, Bak, caspases, pAMPK, pAkt, beclin, p62 and LC3BII) involved in these processes was observed. The results obtained confirmed an important anticancer effect of C. guianensis bark decoction, providing scientific validation for its use in traditional medicine and highlighting its potential as a therapeutic agent against gastric cancer.


Subject(s)
Apoptosis , Cell Proliferation , Plant Bark , Plant Extracts , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Line, Tumor , Plant Bark/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Survival/drug effects , Cell Cycle/drug effects , Autophagy/drug effects
8.
Int J Mol Sci ; 25(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273277

ABSTRACT

Our study highlights the apoptosis, cell cycle, DNA ploidy, and autophagy molecular mechanisms network to identify prostate pathogenesis and its prognostic role. Caspase 3/7 expressions, cell cycle, adhesion glycoproteins, autophagy, nuclear shrinkage, and oxidative stress by flow-cytometry analysis are used to study the BPH microenvironment's heterogeneity. A high late apoptosis expression by caspases 3/7 activity represents an unfavorable prognostic biomarker, a dependent predictor factor for cell adhesion, growth inhibition by arrest in the G2/M phase, and oxidative stress processes network. The heterogeneous aggressive phenotype prostate adenoma primary cell cultures present a high S-phase category (>12%), with an increased risk of death or recurrence due to aneuploid status presence, representing an unfavorable prognostic biomarker, a dependent predictor factor for caspase 3/7 activity (late apoptosis and necrosis), and cell growth inhibition (G2/M arrest)-linked mechanisms. Increased integrin levels in heterogenous BPH cultures suggest epithelial-mesenchymal transition (EMT) that maintains an aggressive phenotype by escaping cell apoptosis, leading to the cell proliferation necessary in prostate cancer (PCa) development. As predictor biomarkers, the biological mechanisms network involved in apoptosis, the cell cycle, and autophagy help to establish patient prognostic survival or target cancer therapy development.


Subject(s)
Apoptosis , Autophagy , Cell Cycle , Prostatic Hyperplasia , Humans , Male , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/genetics , Prognosis , Primary Cell Culture , Epithelial-Mesenchymal Transition/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Phenotype , Aged , Caspase 3/metabolism , Cell Proliferation , Caspase 7/metabolism , Middle Aged , Oxidative Stress
9.
Int J Mol Sci ; 25(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273527

ABSTRACT

NME6 belongs to the family of nucleoside diphosphate kinase enzymes, whose major role is to transfer the terminal phosphate from NTPs, mostly ATP, to other (d)NDPs via a high-energy intermediate. Beside this basic enzymatic activity, the family, comprising 10 genes/proteins in humans, executes a number of diverse biochemical/biological functions in the cell. A few previous studies have reported that NME6 resides in the mitochondria and influences oxidative phosphorylation while interacting with RCC1L, a GTPase involved in mitochondrial ribosome assembly and translation. Considering the multifunctional role of NME family members, the goal of the present study was to assess the influence of the overexpression or silencing of NME6 on fundamental cellular events of MDA-MB-231T metastatic breast cancer cells. Using flow cytometry, Western blotting, and a wound-healing assay, we demonstrated that the overexpression of NME6 reduces cell migration and alters the expression of EMT (epithelial-mesenchymal transition) markers. In addition, NME6 overexpression influences cell cycle distribution exclusively upon DNA damage and impacts the MAPK/ERK signaling pathway, while it has no effect on apoptosis. To conclude, our results demonstrate that NME6 is involved in different cellular processes, providing a solid basis for future, more precise investigations of its role.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Mitochondria , Humans , Cell Line, Tumor , Mitochondria/metabolism , Mitochondria/genetics , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Apoptosis/genetics , Nucleoside Diphosphate Kinase D/metabolism , Nucleoside Diphosphate Kinase D/genetics , Female , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Cell Cycle/genetics
10.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273571

ABSTRACT

Amyloid-beta peptide (Aß) is a neurotoxic constituent of senile plaques in the brains of Alzheimer's disease (AD) patients. The detailed mechanisms by which protein kinase C-delta (PKCδ) contributes to Aß toxicity is not yet entirely understood. Using fully differentiated primary rat cortical neurons, we found that inhibition of Aß25-35-induced PKCδ increased cell viability with restoration of neuronal morphology. Using cyclin D1, proliferating cell nuclear antigen (PCNA), and histone H3 phosphorylated at Ser-10 (p-Histone H3) as the respective markers for the G1-, S-, and G2/M-phases, PKCδ inhibition mitigated cell cycle reentry (CCR) and subsequent caspase-3 cleavage induced by both Aß25-35 and Aß1-42 in the post-mitotic cortical neurons. Upstream of PKCδ, signal transducers and activators of transcription (STAT)-3 mediated PKCδ induction, CCR, and caspase-3 cleavage upon Aß exposure. Downstream of PKCδ, aberrant neuronal CCR was triggered by overactivating cyclin-dependent kinase-5 (CDK5) via calpain2-dependent p35 cleavage into p25. Finally, PKCδ and CDK5 also contributed to Aß25-35 induction of p53-upregulated modulator of apoptosis (PUMA) in cortical neurons. Together, we demonstrated that, in the post-mitotic neurons exposed to Aßs, STAT3-dependent PKCδ expression triggers calpain2-mediated p35 cleavage into p25 to overactivate CDK5, thus leading to aberrant CCR, PUMA induction, caspase-3 cleavage, and ultimately apoptosis.


Subject(s)
Amyloid beta-Peptides , Apoptosis , Cell Cycle , Cerebral Cortex , Neurons , Protein Kinase C-delta , Amyloid beta-Peptides/metabolism , Animals , Neurons/metabolism , Neurons/drug effects , Apoptosis/drug effects , Rats , Protein Kinase C-delta/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Cell Cycle/drug effects , Cyclin-Dependent Kinase 5/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Caspase 3/metabolism , Rats, Sprague-Dawley , Cells, Cultured , Signal Transduction/drug effects
11.
J Toxicol Environ Health A ; 87(22): 895-909, 2024 Nov 16.
Article in English | MEDLINE | ID: mdl-39225403

ABSTRACT

Humans have been using plants in the treatment of various diseases for millennia. Currently, even with allopathic medicines available, numerous populations globally still use plants for therapeutic purposes. Although plants constitute a safer alternative compared to synthetic agents, it is well established that medicinal plants might also exert adverse effects. Thus, the present investigation aimed to assess the phytotoxic, cytotoxic, and genotoxic potential of two plants from the Brazilian Cerrado used in popular medicine, Davilla nitida (Vahl) Kubitzki, and Davilla elliptica (A. St.-Hil.). To this end, germination, growth, and cell cycle analyses were conducted using the plant model Lactuca sativa. Seeds and roots were treated with 0.0625 to 1 g/L for 48 hr under controlled conditions. The germination test demonstrated significant phytotoxic effects for both species at the highest concentrations tested, while none of the extracts produced significant effects in the lettuce growth test. In the microscopic analyses, the aneugenic and cytotoxic action of D. elliptica was evident. In the case of D. nitida greater clastogenic action and induction of micronuclei, (MN) were noted suggesting that the damage initiated by exposure to these extracts was not repaired or led to apoptosis. These findings indicated that the observed plant damage was transmitted to the next generation of cells by way of MN. These differences in the action of the two species may not be attributed to qualitative variations in the composition of the extracts as both are similar, but to quantitative differences associated with synergistic and antagonistic interactions between the compounds present in these extracts.


Subject(s)
Dilleniaceae , Lactuca , Plant Extracts , Plants, Medicinal , Plants, Medicinal/toxicity , Plants, Medicinal/chemistry , Plant Extracts/toxicity , Lactuca/drug effects , Lactuca/growth & development , Dilleniaceae/chemistry , Germination/drug effects , Seeds/drug effects , Brazil , Cell Cycle/drug effects , Plant Roots/drug effects
12.
Asian Pac J Cancer Prev ; 25(9): 3219-3228, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39342601

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma is ranked as the predominant type of head and neck squamous cell carcinoma, comprising roughly 90% of all oral cancer cases. Natural products have proven to be highly valuable as complementary, or adjunctive in the treatment of cancer. Piperine, a natural compound derived from Piper nigrum, demonstrates anti-proliferative and anti-neoplastic effects across various types of cancer. This study focused on assessing the cytotoxic effect of piperine in conjunction with cisplatin within the OSCC cell line. METHODS: In this in-vitro study, cultured OSCC cells were divided into four groups: a control group (untreated), a group exposed solely to piperine, a group exposed solely to cisplatin, and a group receiving both piperine and cisplatin. Cell viability was determined by the 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyltetrazolium bromide (MTT) assay technique. Additionally, flow cytometric analysis was employed to examine cell cycle progression and apoptosis. Assessment of reactive oxygen species activity, morphological changes, and nuclear area factor measurements were carried out. Expression of the apoptotic regulator Bax was assessed through western blotting analysis. RESULTS: Piperine has cytotoxic and apoptotic effects in a concentration-dependent manner. Piperine in combination with cisplatin exhibited a synergistic effect, resulting in more pronounced inhibition of cell viability in OSCC cells compared to using piperine and cisplatin alone. Piperine and cisplatin for 24 h induced apoptosis strongly by increasing Bax protein and ROS activity. CONCLUSION: Combining piperine with cisplatin demonstrated a greater effectiveness in triggering apoptosis in OSCC cells compared to using cisplatin alone, allowing for a reduction.


Subject(s)
Alkaloids , Apoptosis , Benzodioxoles , Carcinoma, Squamous Cell , Cell Proliferation , Cisplatin , Piperidines , Polyunsaturated Alkamides , Tongue Neoplasms , Humans , Polyunsaturated Alkamides/pharmacology , Alkaloids/pharmacology , Piperidines/pharmacology , Benzodioxoles/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Tongue Neoplasms/drug therapy , Tongue Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Drug Synergism , Tumor Cells, Cultured , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cell Cycle/drug effects , Cell Line, Tumor
13.
Asian Pac J Cancer Prev ; 25(9): 3293-3300, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39342609

ABSTRACT

OBJECTIVE: This study aimed to assess linagliptin's inhibitory effects on the proliferation of cervical cancer cell lines and investigate its potential for targeting human heat shock protein 90. METHODS: Linagliptin's cytotoxicity was assessed on a cervical cancer cell line (Hela cancer cell line) at two different incubation periods, 24 and 72 hours. The molecular docking between linagliptin and the receptor protein human Hsp 90 (PDB code: 5XRE) was performed using the Biovia Discovery Studio and AutoDock tool software. The Discovery Studio visualizer generated three-dimensional (3D) and two-dimensional (2D) interactive images. RESULTS: The study's cytotoxicity results demonstrated that linagliptin can inhibit the proliferation of cervical cancer cells. The cytotoxicity exhibited a time-dependent pattern (cell cycle specific). The molecular docking study was conducted to investigate the interaction between linagliptin and human Hsp90. The study identified 11 sites where linagliptin can bind to Hsp90 amino acid residues. The total docking score for this interaction was -10.3 kcal/mol. The most potent binding occurred through conventional hydrogen bonds with the ASP:54 amino acid residues at a distance of 2.93 Å. The docking scores for linagliptin were comparable to those of the reference drug geldanamycin, indicating a strong interaction between linagliptin and Hsp90. CONCLUSION: The study has found that linagliptin successfully reduces the growth of cervical cancer cells with a time-dependent cytotoxic pattern. The potential anticancer mechanism of linagliptin can be inferred by analyzing the docking score and docking pattern between linagliptin and Hsp90, suggesting that linagliptin targets human Hsp 90.


Subject(s)
Cell Proliferation , HSP90 Heat-Shock Proteins , Linagliptin , Molecular Docking Simulation , Uterine Cervical Neoplasms , Humans , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Linagliptin/pharmacology , Cell Proliferation/drug effects , Female , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Tumor Cells, Cultured , Cell Cycle/drug effects , Apoptosis/drug effects , HeLa Cells
14.
Biomolecules ; 14(9)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39334853

ABSTRACT

Breast cancer (BC) is the most frequently diagnosed cancer and the primary cause of cancer-related mortality in women. Treatment of triple-negative breast cancer (TNBC) remains particularly challenging due to its resistance to chemotherapy and poor prognosis. Extensive research efforts in BC screening and therapy have improved clinical outcomes for BC patients. Therefore, identifying reliable biomarkers for TNBC is of great clinical importance. Here, we found that tyrosine aminotransferase (TAT) expression was significantly reduced in BC and strongly correlated with the poor prognosis of BC patients, which distinguished BC patients from normal individuals, indicating that TAT is a valuable biomarker for early BC diagnosis. Mechanistically, we uncovered that methylation of the TAT promoter was significantly increased by DNA methyltransferase 3 (DNMT3A/3B). In addition, reduced TAT contributes to DNA replication and cell cycle activation by regulating homologous recombination repair and mismatch repair to ensure genomic stability, which may be one of the reasons for TNBC resistance to chemotherapy. Furthermore, we demonstrated that Diazinon increases TAT expression as an inhibitor of DNMT3A/3B and inhibits the growth of BC by blocking downstream pathways. Taken together, we revealed that TAT is silenced by DNMT3A/3B in BC, especially in TNBC, which promotes the proliferation of tumor cells by supporting DNA replication, activating cell cycle, and enhancing DNA damage repair. These results provide fresh insights and a theoretical foundation for the clinical diagnosis and treatment of BC.


Subject(s)
Biomarkers, Tumor , Cell Cycle , DNA Methyltransferase 3A , DNA Repair , Triple Negative Breast Neoplasms , Humans , Female , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , DNA Repair/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , DNA Methyltransferase 3A/metabolism , Cell Cycle/genetics , Cell Line, Tumor , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Methyltransferase 3B , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Promoter Regions, Genetic/genetics , Prognosis
15.
Molecules ; 29(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39339471

ABSTRACT

Lithium, a natural element, has been employed as a mental stabilizer in psychiatric treatments; however, some reports indicate it has an anticancer effect, prompting the consideration of repurposing lithium for cancer treatment. The potential anticancer use of lithium may depend on its form (salt type) and the type of cancer cells targeted. Little is known about the effects of Li2CO3 or LiCl on cancer cells, so we focused on exploring their effects on proliferation, apoptosis, migration, and cell cycle as part of the hallmarks of cancer. Firstly, we established the IC50 values on HeLa, SiHa, and HaCaT cells with LiCl and Li2CO3 and determined by crystal violet that cell proliferation was time-dependent in the three cell lines (IC50 values for LiCl were 23.43 mM for SiHa, 23.14 mM for HeLa, and 15.10 mM for HaCaT cells, while the IC50 values for Li2CO3 were 20.57 mM for SiHa, 11.52 mM for HeLa, and 10.52 mM for HaCaT cells.) Our findings indicate that Li2CO3 and LiCl induce DNA fragmentation and caspase-independent apoptosis, as shown by TUNEL, Western Blot, and Annexin V/IP assay by flow cytometry. Also, cell cycle analysis showed that LiCl and Li2CO3 arrested the cervical cancer cells at the G1 phase. Moreover, lithium salts displayed an anti-migratory effect on the three cell lines observed by the wound-healing assay. All these findings imply the viable anticancer effect of lithium salts by targeting several of the hallmarks of cancer.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Lithium Chloride , Uterine Cervical Neoplasms , Humans , Lithium Chloride/pharmacology , Cell Proliferation/drug effects , Apoptosis/drug effects , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Cell Movement/drug effects , Female , HeLa Cells , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Lithium Carbonate/pharmacology , Cell Cycle/drug effects , Drug Repositioning
16.
FASEB J ; 38(18): e70058, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39320969

ABSTRACT

Uric acid (UA) is the end product of purine metabolism. In recent years, UA has been found to be associated with the prognosis of clinical cancer patients. However, the intricate mechanisms by which UA affects the development and prognosis of tumor patients has not been well elucidated. In this study, we explored the role of UA in breast cancer, scrutinizing its impact on breast cancer cell function by treating two types of breast cancer cell lines with UA. The role of UA in the cell cycle and proliferation of tumors and the underlying mechanisms were further investigated. We found that the antioxidant effect of UA facilitated the scavenging of reactive oxygen species (ROS) in breast cancer, thereby reducing aryl hydrocarbon receptor (AhR) expression and affecting the breast cancer cell cycle, driving the proliferation of breast cancer cells through the AhR/p27Kip1/cyclin E1 pathway. Moreover, in breast cancer patients, the expression of AhR and its downstream genes may be closely associated with cancer progression in patients. Therefore, an increase in UA could promote the proliferation of breast cancer cells through the AhR/p27Kip1/cyclin E1 pathway axis.


Subject(s)
Breast Neoplasms , Cell Proliferation , Cyclin E , Cyclin-Dependent Kinase Inhibitor p27 , Receptors, Aryl Hydrocarbon , Humans , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Cyclin E/metabolism , Cyclin E/genetics , Cell Line, Tumor , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Signal Transduction , Reactive Oxygen Species/metabolism , Disease Progression , Animals , Oncogene Proteins/metabolism , Oncogene Proteins/genetics , Mice , Gene Expression Regulation, Neoplastic , Cell Cycle , Mice, Nude
17.
PLoS Pathog ; 20(9): e1012535, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39255317

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is a double-stranded DNA virus that encodes numerous cellular homologs, including cyclin D, G protein-coupled protein, interleukin-6, and macrophage inflammatory proteins 1 and 2. KSHV vCyclin encoded by ORF72, is the homolog of cellular cyclinD2. KSHV vCyclin can regulate virus replication and cell proliferation by constitutively activating cellular cyclin-dependent kinase 6 (CDK6). However, the regulatory mechanism of KSHV vCyclin has not been fully elucidated. In the present study, we identified a host protein named protein arginine methyltransferase 5 (PRMT5) that interacts with KSHV vCyclin. We further demonstrated that PRMT5 is upregulated by latency-associated nuclear antigen (LANA) through transcriptional activation. Remarkably, knockdown or pharmaceutical inhibition (using EPZ015666) of PRMT5 inhibited the cell cycle progression and cell proliferation of KSHV latently infected tumor cells. Mechanistically, PRMT5 methylates vCyclin symmetrically at arginine 128 and stabilizes vCyclin in a methyltransferase activity-dependent manner. We also show that the methylation of vCyclin by PRMT5 positively regulates the phosphorylate retinoblastoma protein (pRB) pathway. Taken together, our findings reveal an important regulatory effect of PRMT5 on vCyclin that facilitates cell cycle progression and proliferation, which provides a potential therapeutic target for KSHV-associated malignancies.


Subject(s)
Cell Cycle , Cell Proliferation , Herpesvirus 8, Human , Protein-Arginine N-Methyltransferases , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Humans , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/physiology , Methylation , Antigens, Viral/metabolism , Antigens, Viral/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Cyclin D2/metabolism , HEK293 Cells , Virus Replication/physiology , Sarcoma, Kaposi/virology , Sarcoma, Kaposi/metabolism , Nuclear Proteins
18.
Medicine (Baltimore) ; 103(38): e39752, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39312302

ABSTRACT

Poria cocos, also known as Jade Ling and Songbai taro, is a dry fungus core for Wolfiporia cocos, which is parasitic on the roots of pine trees. The ancients called it "medicine of four seasons" because of its extensive effect and ability to be combined with many medicines. Pachymic acid (PA) is one of the main biological compounds of Poria cocos. Research has shown that PA has various pharmacological properties, including anti-inflammatory and antioxidant. PA has recently attracted much attention due to its anticancer properties. Researchers have found that PA showed anticancer activity by regulating apoptosis and the cell cycle in vitro and in vivo. Using PA with anticancer drugs, radiotherapy, and biomaterials could also improve the sensitivity of cancer cells and delay the progression of cancer. The purpose of this review was to summarize the anticancer mechanism of PA by referencing the published documents. A review of the collected data indicated that PA had the potential to be developed into an effective anticancer agent.


Subject(s)
Antineoplastic Agents , Triterpenes , Triterpenes/pharmacology , Triterpenes/therapeutic use , Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Animals , Wolfiporia/chemistry , Cell Cycle/drug effects
19.
Medicine (Baltimore) ; 103(38): e39658, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39312343

ABSTRACT

Lung cancer is a common malignant tumor with low cure rate. It has an easy recurrence and metastasis. This study explored whether miR-200c could regulate the biological behavior of non-small cell lung cancer cells through targeting GLI3. Luciferase reporter gene analysis was used to verify the interaction between miR-200c-3p and GLI3. miR-200c-3p and GLI3 were transiently overexpressed into A549 cells. The cell viability rate was detected by cell counting kit-8, cell invasion ability was detected with Transwell, cell apoptosis and cell cycle was determined by flow cytometry, and the expression of GLI3 was detected using quantitative polymerase chain reaction and Western blot, to verify the effect of the interaction between miR-200c-3p and GLI3 on the cell activities. miR-200c-3p overexpression could inhibit cell viability and invasion, promote apoptosis, induce G0/G1 arrest, and inhibit cell division. GLI3 overexpression could reverse the miR-200c-3p inhibition on cell cycle, reduce the number of cells in the G0/G1 phase and increase the number of cells in the S phase. miR-200c-3p overexpression in A549 cells could inhibit cell viability and invasion, and promote apoptosis. miR-200c-3p could target GLI3 to regulate cell cycle and inhibit cell proliferation.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung , Cell Proliferation , Lung Neoplasms , MicroRNAs , Zinc Finger Protein Gli3 , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Apoptosis/genetics , Cell Proliferation/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , A549 Cells , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Cell Survival/genetics , Cell Cycle
20.
Sci Rep ; 14(1): 19155, 2024 09 02.
Article in English | MEDLINE | ID: mdl-39223145

ABSTRACT

Traditional chemotherapy drugs for cervical cancer often cause significant toxic side effects and drug resistance problems, highlighting the urgent need for more innovative and effective treatment strategies. Magnesium alloy is known to be degradable and biocompatible. The release of degradation products Mg2+, OH-, and H2 from magnesium alloy can alter the tumor microenvironment, providing potential anti-tumor properties. We explored the innovative use of magnesium alloy biomaterials in the treatment of cervical cancer, investigating how various concentrations of Mg2+ on the proliferation and cell death of cervical cancer cells. The results revealed that varying concentrations of Mg2+ significantly inhibited cervical cancer by arresting the cell cycle in the G0/G1 phase and inducing apoptosis in SiHa cells, effectively reducing tumor cell proliferation. In vivo experiments demonstrated that 20 mM Mg2+ group had the smallest tumor volume, exhibiting a potent inhibitory effect on the biological characteristics of cervical cancer. This enhances the therapeutic potential of this biomaterial as a local anti-tumor therapy and lays a theoretical foundation for the potential application of magnesium in the treatment of cervical cancer.


Subject(s)
Apoptosis , Biocompatible Materials , Cell Proliferation , Magnesium , Uterine Cervical Neoplasms , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Female , Magnesium/pharmacology , Magnesium/chemistry , Humans , Cell Proliferation/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Alloys/pharmacology , Alloys/chemistry , Xenograft Model Antitumor Assays , Cell Cycle/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL