Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66.236
Filter
1.
Cell Signal ; 119: 111172, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38604342

ABSTRACT

Simvastatin is an inhibitor of 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, which is a rate-limiting enzyme of the cholesterol synthesis pathway. It has been used clinically as a lipid-lowering agent to reduce low-density lipoprotein (LDL) cholesterol levels. In addition, antitumor activity has been demonstrated. Although simvastatin attenuates the prenylation of small GTPases, its effects on cell division in which small GTPases play an important role, have not been examined as a mechanism underlying its cytostatic effects. In this study, we determined its effect on cell division. Cell cycle synchronization experiments revealed a delay in mitotic progression in simvastatin-treated cells at concentrations lower than the IC50. Time-lapse imaging analysis indicated that the duration of mitosis, especially from mitotic entry to anaphase onset, was prolonged. In addition, simvastatin increased the number of cells exhibiting misoriented anaphase/telophase and bleb formation. Inhibition of the spindle assembly checkpoint (SAC) kinase Mps1 canceled the mitotic delay. Additionally, the number of cells exhibiting kinetochore localization of BubR1, an essential component of SAC, was increased, suggesting an involvement of SAC in the mitotic delay. Enhancement of F-actin formation and cell rounding at mitotic entry indicates that cortical actin dynamics were affected by simvastatin. The cholesterol removal agent methyl-ß-cyclodextrin (MßCD) accelerated mitotic progression differently from simvastatin, suggesting that cholesterol loss from the plasma membrane is not involved in the mitotic delay. Of note, the small GTPase RhoA, which is a critical factor for cortical actin dynamics, exhibited upregulated expression. In addition, Rap1 was likely not geranylgeranylated. Our results demonstrate that simvastatin affects actin dynamics by modifying small GTPases, thereby activating the spindle assembly checkpoint and causing abnormal cell division.


Subject(s)
M Phase Cell Cycle Checkpoints , Simvastatin , Simvastatin/pharmacology , Humans , M Phase Cell Cycle Checkpoints/drug effects , HeLa Cells , Monomeric GTP-Binding Proteins/metabolism , Mitosis/drug effects , Cell Division/drug effects , rhoA GTP-Binding Protein/metabolism
2.
Sci Rep ; 14(1): 9029, 2024 04 19.
Article in English | MEDLINE | ID: mdl-38641657

ABSTRACT

Double-stranded RNA-activated protein kinase R (PKR) is highly expressed in colorectal cancer (CRC). However, the role of PKR in CRC remains unclear. The aim of this study was to clarify whether C16 (a PKR inhibitor) exhibits antitumor effects and to identify its target pathway in CRC. We evaluated the effects of C16 on CRC cell lines using the MTS assay. Enrichment analysis was performed to identify the target pathway of C16. The cell cycle was analyzed using flow cytometry. Finally, we used immunohistochemistry to examine human CRC specimens. C16 suppressed the proliferation of CRC cells. Gene Ontology (GO) analysis revealed that the cell cycle-related GO category was substantially enriched in CRC cells treated with C16. C16 treatment resulted in G1 arrest and increased p21 protein and mRNA expression. Moreover, p21 expression was associated with CRC development as observed using immunohistochemical analysis of human CRC tissues. C16 upregulates p21 expression in CRC cells to regulate cell cycle and suppress tumor growth. Thus, PKR inhibitors may serve as a new treatment option for patients with CRC.


Subject(s)
Colorectal Neoplasms , Protein Kinase Inhibitors , Humans , Apoptosis , Cell Cycle , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Protein Kinase Inhibitors/pharmacology , Indoles/pharmacology , Thiazoles/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism
3.
Chemosphere ; 358: 142125, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670509

ABSTRACT

Microcystin-LR (MC-LR) is a harmful cyanotoxin that inhibits 1 and 2A serine-threonine protein phosphatases. This study examines the influence of MC-LR on chloroplast division and the underlying mechanisms and consequences in Arabidopsis. MC-LR increased the frequency of dividing chloroplasts in hypocotyls in a time range of 1-96 h. At short-term exposures to MC-LR, small-sized chloroplasts (longitudinal diameters ≤6 µm) were more sensitive to these stimulatory effects, while both small and large chloroplasts showed stimulations at long-term exposure. After 48 h, the cyanotoxin increased the frequency of small-sized chloroplasts, indicating the stimulation of division. MC-LR inhibited protein phosphatases in whole hypocotyls and isolated chloroplasts, while it did not induce oxidative stress. We show for the first time that total cellular phosphatases play important roles in chloroplast division and that particular chloroplast phosphatases may be involved in these processes. Interestingly, MC-LR has a protective effect on cyanobacterial division during methyl-viologen (MV) treatments in Synechococcus PCC6301. MC-LR production has harmful effects on ecosystems and it may have an ancient cell division regulatory role in stressed cyanobacterial cells, the evolutionary ancestors of chloroplasts. We propose that cytoplasmic (eukaryotic) factors also contribute to the relevant effects of MC-LR in plants.


Subject(s)
Arabidopsis , Chloroplasts , Marine Toxins , Microcystins , Phosphoprotein Phosphatases , Microcystins/toxicity , Chloroplasts/drug effects , Chloroplasts/metabolism , Phosphoprotein Phosphatases/metabolism , Arabidopsis/drug effects , Cyanobacteria/drug effects , Cell Division/drug effects , Synechococcus/drug effects
4.
J Biophotonics ; 17(5): e202300369, 2024 May.
Article in English | MEDLINE | ID: mdl-38332564

ABSTRACT

5-Aminolevulinic acid (5-ALA) photodynamic therapy (PDT) is a treatment for actinic keratosis (AK) and has been studied as a treatment for noninvasive cutaneous squamous cell carcinoma (cSCC). PDT induces apoptosis and necrosis in AKs and cSCC. 5-ALA blue light PDT may modulate gene expression and pathways in surviving cells. In this study, differential gene expression and pathway analysis of cSCC and human dermal fibroblasts were compared before and after 5-ALA blue light PDT using RNA sequencing. No genes were differentially expressed after correcting for multiple testing (false discovery rate < 0.05). As a result, transcription factor, gene enrichment, and pathway analysis were performed with genes identified before multiple testing (p < 0.05). Pathways associated with proliferation and carcinogenesis were downregulated. These findings using 5-ALA blue light PDT are similar to previously published studies using methyl-aminolevulinic and red light protocols, indicating that surviving residual cells may undergo changes consistent with a less aggressive cancerous phenotype.


Subject(s)
Aminolevulinic Acid , Carcinoma, Squamous Cell , Cell Proliferation , Down-Regulation , Photochemotherapy , Skin Neoplasms , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Humans , Skin Neoplasms/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/drug effects , Down-Regulation/drug effects , Cell Division/drug effects , Cell Division/radiation effects , Light , Gene Expression Regulation, Neoplastic/drug effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Blue Light
5.
Curr Drug Targets ; 25(3): 190-200, 2024.
Article in English | MEDLINE | ID: mdl-38213162

ABSTRACT

Physiologically, cytokines play an extremely important role in maintaining cellular and subcellular homeostasis, as they interact almost with every cell in the organism. Therefore, cytokines play a significantly critical role in the field of pathogenic pharmacological therapy of different types of pathologies. Cytokine is a large family containing many subfamilies and can be evaluated into groups according to their action on epithelial cell proliferation; stimulatory include transforming growth factor-α (TGF-α), Interlukine-22 (IL-22), IL-13, IL-6, IL-1RA and IL-17 and inhibitory include IL-1α, interferon type I (IFN type I), and TGF-ß. The balance between stimulatory and inhibitory cytokines is essential for maintaining normal epithelial cell turnover and tissue homeostasis. Dysregulation of cytokine production can contribute to various pathological conditions, including inflammatory disorders, tissue damage, and cancer. Several cytokines have shown the ability to affect programmed cell death (apoptosis) and the capability to suppress non-purpose cell proliferation. Clinically, understanding the role of cytokines' role in epithelial tissue is crucial for evaluating a novel therapeutic target that can be of use as a new tactic in the management of carcinomas and tissue healing capacity. The review provides a comprehensive and up-to-date synthesis of current knowledge regarding the multifaceted effects of cytokines on epithelial cell proliferation, with a particular emphasis on the intestinal epithelium. Also, the paper will highlight the diverse signaling pathways activated by cytokines and their downstream consequences on epithelial cell division. It will also explore the potential therapeutic implications of targeting cytokine- epithelial cell interactions in the context of various diseases.


Subject(s)
Cell Proliferation , Cytokines , Epithelial Cells , Humans , Cytokines/metabolism , Epithelial Cells/metabolism , Animals , Cell Division/drug effects , Signal Transduction , Apoptosis/drug effects , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/drug therapy
6.
Nature ; 619(7968): 160-166, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37258666

ABSTRACT

KRAS is one of the most commonly mutated proteins in cancer, and efforts to directly inhibit its function have been continuing for decades. The most successful of these has been the development of covalent allele-specific inhibitors that trap KRAS G12C in its inactive conformation and suppress tumour growth in patients1-7. Whether inactive-state selective inhibition can be used to therapeutically target non-G12C KRAS mutants remains under investigation. Here we report the discovery and characterization of a non-covalent inhibitor that binds preferentially and with high affinity to the inactive state of KRAS while sparing NRAS and HRAS. Although limited to only a few amino acids, the evolutionary divergence in the GTPase domain of RAS isoforms was sufficient to impart orthosteric and allosteric constraints for KRAS selectivity. The inhibitor blocked nucleotide exchange to prevent the activation of wild-type KRAS and a broad range of KRAS mutants, including G12A/C/D/F/V/S, G13C/D, V14I, L19F, Q22K, D33E, Q61H, K117N and A146V/T. Inhibition of downstream signalling and proliferation was restricted to cancer cells harbouring mutant KRAS, and drug treatment suppressed KRAS mutant tumour growth in mice, without having a detrimental effect on animal weight. Our study suggests that most KRAS oncoproteins cycle between an active state and an inactive state in cancer cells and are dependent on nucleotide exchange for activation. Pan-KRAS inhibitors, such as the one described here, have broad therapeutic implications and merit clinical investigation in patients with KRAS-driven cancers.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Animals , Mice , Body Weight , Enzyme Activation , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Nucleotides/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects , Cell Division/drug effects , Substrate Specificity
7.
Med Oncol ; 39(9): 124, 2022 Jun 18.
Article in English | MEDLINE | ID: mdl-35716217

ABSTRACT

Both pro-oncogenic and anti-oncogenic effects of E2F2 have been revealed in different malignancies. However, the precise role of E2F2 in pancreatic cancer, in particular in relation to therapeutic intervention with gemcitabine, remains unclear. In this study, the effect of E2F2 on the proliferation and cell cycle modulation of pancreatic cancer cells, and whether E2F2 plays a role in the treatment of pancreatic cancer cells by gemcitabine, were investigated. The expression of E2F2 in pancreatic cancer was assessed by various methods including bioinformatics prediction, Western blotting, and real-time PCR. The effect of E2F2 on the proliferation and cell cycling of pancreatic cancer cells was analyzed by tissue culture and flow cytometry. In addition, the effect of E2F2 on the intervention of pancreatic cancer by gemcitabine was investigated using both in vitro and in vivo approaches. The expression of E2F2 was found to be significantly increased in pancreatic cancer tissues and cell lines. The pathogenic capacity of E2F2 lied in the fact that this transcription factor promoted the transformation of pancreatic cancer cell cycle from G1-phase to S-phase, thus enhancing the proliferation of pancreatic cancer cells. Furthermore, the expression of E2F2 was increased in pancreatic cancer cells in the presence of gemcitabine, and the augmented expression of E2F2 upregulated the gemcitabine resistance-related gene RRM2 and its downstream signaling molecule deoxycytidine kinase (DCK). The resistance of pancreatic cancer cells to gemcitabine was confirmed using both in vitro and in vivo models. In this study, E2F2 has been demonstrated for the first time to play a pro-oncogenic role in pancreatic cancer by promoting the transition of the cell cycle from G1-phase to S-phase and, therefore, enhancing the proliferation of pancreatic cancer cells. E2F2 has also been demonstrated to enhance the chemotherapy resistance of pancreatic cancer cells to gemcitabine by upregulating the expression of RRM2 and DCK that is downstream of RRM2.


Subject(s)
Deoxycytidine , Drug Resistance, Neoplasm , E2F2 Transcription Factor , Pancreatic Neoplasms , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Ribonucleoside Diphosphate Reductase/biosynthesis , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Up-Regulation/drug effects , Gemcitabine
8.
J Med Chem ; 65(4): 3644-3666, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35119851

ABSTRACT

We recently reported a potent, selective, and in vivo efficacious AKT degrader, MS21, which is a von Hippel-Lindau (VHL)-recruiting proteolysis targeting chimera (PROTAC) based on the AKT inhibitor AZD5363. However, no structure-activity relationship (SAR) studies that resulted in this discovery have been reported. Herein, we present our SAR studies that led to the discovery of MS21, another VHL-recruiting AKT degrader, MS143 (compound 20) with similar potency as MS21, and a novel cereblon (CRBN)-recruiting PROTAC, MS5033 (compound 35). Compounds 20 and 35 induced rapid and robust AKT degradation in a concentration- and time-dependent manner via hijacking the ubiquitin-proteasome system. Compound 20 suppressed cell growth more effectively than AZD5363 in multiple cancer cell lines. Furthermore, 20 and 35 displayed good plasma exposure levels in mice and are suitable for in vivo efficacy studies. Lastly, compound 20 effectively suppressed tumor growth in vivo in a xenograft model without apparent toxicity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Cell Division/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Nude , PC-3 Cells , Proteasome Endopeptidase Complex/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Proteolysis , Proto-Oncogene Proteins c-akt/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Structure-Activity Relationship , Tumor Stem Cell Assay , Ubiquitin/genetics , Xenograft Model Antitumor Assays
10.
Plant Sci ; 314: 111117, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34895546

ABSTRACT

The MEDIATOR complex influences the transcription of genes acting as a RNA pol II co-activator. The MED16 subunit has been related to low phosphate sensing in roots, but how it influences the overall plant growth and root development remains unknown. In this study, we compared the root growth of Arabidopsis wild-type (WT), and two alleles of MED16 (med16-2 and med16-3) mutants in vitro. The MED16 loss-of-function seedlings showed longer primary roots with higher cell division capacity of meristematic cells, and an increased number of lateral roots than WT plants, which correlated with improved biomass accumulation. The auxin response reported by DR5:GFP fluorescence was comparable in WT and med16-2 root tips, but strongly decreased in pericycle cells and lateral root primordia in the mutants. Dose-response analysis supplementing indole-3-acetic acid (IAA), or the auxin transport inhibitor N-1-naphthylphthalamic acid (NPA), indicated normal responses to auxin in the med16-2 and med16-3 mutants regarding primary root growth and lateral root formation, but strong resistance to NPA in primary roots, which could be correlated with cell division and elongation. Expression analysis of pPIN1::PIN1::GFP, pPIN3::PIN3::GFP, pIAA14:GUS, pIAA28:GUS and 35S:MED16-GFP suggests that MED16 could mediate auxin signaling. Our data imply that an altered auxin response in the med16 mutants is not necessarily deleterious for overall growth and developmental patterning and may instead directly regulate basic cellular programmes.


Subject(s)
Arabidopsis/growth & development , Arabidopsis/genetics , Arabidopsis/metabolism , Indoleacetic Acids/metabolism , Plant Roots/growth & development , Plant Roots/genetics , Plant Roots/metabolism , Signal Transduction/drug effects , Biomass , Cell Division/drug effects , Cell Division/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Plant , Genes, Plant , Genetic Variation , Mutation
11.
Eur J Med Chem ; 229: 114002, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34823899

ABSTRACT

Compounds targeting the inflammasome-caspase-1 pathway could be of use for the treatment of inflammation and inflammatory diseases. Previous caspase-1 inhibitors were in great majority covalent inhibitors and failed in clinical trials. Using a mixed modelling, computational screening, synthesis and in vitro testing approach, we identified a novel class of non-covalent caspase-1 non cytotoxic inhibitors which are able to inhibit IL-1ß release in activated macrophages in the low µM range, in line with the best activities observed for the known covalent inhibitors. Our compounds could form the basis of further optimization towards potent drugs for the treatment of inflammation and inflammatory disorders including also dysregulated inflammation in Covid 19.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Autoimmune Diseases/drug therapy , Caspase 1/drug effects , Inflammasomes/drug effects , Inflammation/drug therapy , Serpins/chemical synthesis , Serpins/pharmacology , Tetrazoles/chemical synthesis , Tetrazoles/therapeutic use , Viral Proteins/chemical synthesis , Viral Proteins/pharmacology , COVID-19 , Cell Division/drug effects , Drug Design , Drug Evaluation, Preclinical , Humans , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Tetrazoles/pharmacology , U937 Cells
12.
Biomed Res Int ; 2021: 5013065, 2021.
Article in English | MEDLINE | ID: mdl-34938808

ABSTRACT

Osteosarcoma is considered to be a highly malignant tumor affecting primarily long bones. It metastasizes widely, primarily to the lungs, resulting in poor survival rates of between 19 and 30%. Standard treatment consists of surgical removal of the affected site, with neoadjuvant and adjuvant chemotherapy commonly used, with the usual side effects and complications. There is a need for new treatments in this area, and silver nanoparticles (AgNPs) are one potential avenue for exploration. AgNPs have been found to possess antitumor and cytotoxic activity in vitro, by demonstrating decreased viability of cancer cells through cell cycle arrest and subsequent apoptosis. Integral to these pathways is tumor protein p53, a tumor suppressor which plays a critical role in maintaining genome stability by regulating cell division, after DNA damage. The purpose of this study was to determine if p53 mediates any difference in the response of the osteosarcoma cells in vitro when different sizes and concentrations of AgNPs are administered. Two cell lines were studied: p53-expressing HOS cells and p53-deficient Saos-2 cells. The results of this study suggest that the presence of protein p53 significantly affects the efficacy of AgNPs on osteosarcoma cells.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Metal Nanoparticles/administration & dosage , Osteosarcoma/drug therapy , Silver/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cytotoxins/pharmacology , DNA Damage/drug effects , Humans , Osteosarcoma/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism
13.
Cell Rep ; 37(12): 110129, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34936867

ABSTRACT

Writing and erasing of posttranslational modifications are crucial to phenotypic plasticity and antigenic variation of eukaryotic pathogens. Targeting pathogens' modification machineries, thus, represents a valid approach to fighting parasitic diseases. However, identification of parasitic targets and the development of selective anti-parasitic drugs still represent major bottlenecks. Here, we show that the zinc-dependent histone deacetylases (HDACs) of the protozoan parasite Trypanosoma cruzi are key regulators that have significantly diverged from their human counterparts. Depletion of T. cruzi class I HDACs tcDAC1 and tcDAC2 compromises cell-cycle progression and division, leading to cell death. Notably, tcDAC2 displays a deacetylase activity essential to the parasite and shows major structural differences with human HDACs. Specifically, tcDAC2 harbors a modular active site with a unique subpocket targeted by inhibitors showing substantial anti-parasitic effects in cellulo and in vivo. Thus, the targeting of the many atypical HDACs in pathogens can enable anti-parasitic selective chemical impairment.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/genetics , Trypanosoma cruzi/metabolism , Animals , Catalytic Domain , Cell Cycle , Cell Division/drug effects , Cell Line , Cell Proliferation/drug effects , Chagas Disease/drug therapy , Chagas Disease/parasitology , Chlorocebus aethiops , DNA, Protozoan , Female , Genetic Complementation Test , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Host-Parasite Interactions , Humans , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Phylogeny , Protein Conformation , Protein Processing, Post-Translational , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Sequence Deletion , Trypanosoma cruzi/drug effects , Vero Cells
14.
Cytogenet Genome Res ; 161(10-11): 501-513, 2021.
Article in English | MEDLINE | ID: mdl-34879371

ABSTRACT

MicroRNA-141 (miR-141-3p) is upregulated in preeclampsia. This study investigated the effect of methylation of the miR-141-3p promoter on cell viability, invasion capability, and inflammasomes in vitro. The expression of miR-141-3p and methylation status of the miR-141-3p promoter were examined by RT-qPCR and pyrosequencing in villus tissues of women with spontaneous delivery (VTsd), villus tissues of women with preeclampsia (VTpe), and also in HTR-8/SVneo cells treated with a miR-141-3p inhibitor and 20 µmol/L 5-aza-2'-deoxycytidine (5-Aza), a DNA methyltransferase inhibitor. Cell viability and invasion were evaluated by CCK-8 and transwell assays. In addition, the levels of CXCL12, CXCR4, CXCR2, MMPs, NLRP3, and ASC expression were assessed by western blotting, and IL-1ß and IL-18 concentrations were assayed by ELISA. miR-141-3p expression was upregulated, and the levels of miR-141-3p promoter methylation and CXCL12, CXCR4, and CXCR2 expression were decreased in VTpe relative to VTsd. In HTR-8/SVneo cells, hypomethylation caused by 5-Aza treatment increased miR-141-3p expression, while DNA methyltransferase 3 (DNMT3) transfection decreased miR-141-3p expression. miRNA-141-3p induced NLRP3, IL-1ß, and IL-18 production, decreased CXCR4, MMP, and MMP2 production, and suppressed cell growth and invasion. Furthermore, we observed that NLRP3 plays an important mediatory role in the effects of miR-141-3p described above. Decreased methylation of the miR-141-3p promoter increases miR-141-3p expression, which in turn increases NLRP3 expression, resulting in higher IL-1ß and IL-18 levels and lower levels of MMP2/9 and CXCR4. We conclude that modification of the miR-141-3p promoter might be a curial mediator in preeclampsia.


Subject(s)
DNA Methylation , Inflammasomes/metabolism , MicroRNAs/genetics , Pre-Eclampsia/pathology , Promoter Regions, Genetic/genetics , Cell Division/drug effects , Cell Movement/drug effects , Chorionic Villi/metabolism , Chorionic Villi/pathology , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , Decitabine/pharmacology , Female , Humans , Interleukin-18/analysis , Interleukin-1beta/analysis , Matrix Metalloproteinases/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , DNA Methyltransferase 3B
15.
Biol Pharm Bull ; 44(12): 1878-1885, 2021.
Article in English | MEDLINE | ID: mdl-34853271

ABSTRACT

Renal cell carcinoma (RCC) is the most common type of kidney cancer. Given that stage IV RCC is intractable, there is a need for a novel treatment strategy. We investigated the antitumor effects of telmisartan (TEL) and their underlying mechanisms in RCC, including their impact on apoptosis, Akt/mammalian target of rapamycin (mTOR) pathways, and the cell cycle using two human RCC cell lines: 786-O and Caki-2. Cell viability was detected via fluorescence-based assays. Cells were stained with Hoechst 33342 to observe chromatin condensation, and Western blotting was performed to analyze protein expression. The cell cycle was assessed using flow cytometry. Invasion and migration assays were performed using 24-well chambers. TEL induced cell death in a dose-dependent manner and increased the percentage of cells with high chromatin condensation and Bax/Bcl-2 ratio in both cell lines. TEL-induced cell death was attenuated by neither peroxisome proliferator-activated receptor-γ nor -δ inhibitors. Although TEL elevated c-Jun N-terminal kinase levels and p38 phosphorylation rates in Caki-2 cells, as well as extracellular signal-regulated kinase phosphorylation rates in 786-O cells, their inhibitors did not suppress TEL-induced cell death. TEL decreased Akt phosphorylation in 786-O cells and mTOR phosphorylation in both cell lines, increased the population of cells in the G2/M phase, and altered G2/M-related proteins in both cell lines. TEL moderately suppressed cell invasion and migration in 786-O and Caki-2 cells, respectively, and increased cell invasion in Caki-2 cells, suggesting a potential therapeutic role of TEL in RCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Kidney Neoplasms , Telmisartan/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , G2 Phase Cell Cycle Checkpoints , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/drug effects , Kidney/pathology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , PPAR gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , TOR Serine-Threonine Kinases/metabolism , Telmisartan/therapeutic use , bcl-2-Associated X Protein/metabolism
16.
Nat Commun ; 12(1): 6925, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34836956

ABSTRACT

All females adopt an evolutionary conserved reproduction strategy; under unfavorable conditions such as scarcity of food or mates, oocytes remain quiescent. However, the signals to maintain oocyte quiescence are largely unknown. Here, we report that in four different species - Caenorhabditis elegans, Caenorhabditis remanei, Drosophila melanogaster, and Danio rerio - octopamine and norepinephrine play an essential role in maintaining oocyte quiescence. In the absence of mates, the oocytes of Caenorhabditis mutants lacking octopamine signaling fail to remain quiescent, but continue to divide and become polyploid. Upon starvation, the egg chambers of D. melanogaster mutants lacking octopamine signaling fail to remain at the previtellogenic stage, but grow to full-grown egg chambers. Upon starvation, D. rerio lacking norepinephrine fails to maintain a quiescent primordial follicle and activates an excessive number of primordial follicles. Our study reveals an evolutionarily conserved function of the noradrenergic signal in maintaining quiescent oocytes.


Subject(s)
Cell Division/drug effects , Norepinephrine/pharmacology , Oocytes/drug effects , Animals , Caenorhabditis/genetics , Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Evolution, Molecular , Female , Food , Nutrients , Octopamine/pharmacology , Oocytes/cytology , Oogenesis , Ovarian Follicle/cytology , Ovarian Follicle/physiology , Starvation , Zebrafish/genetics
17.
Biomed Pharmacother ; 144: 112358, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34794241

ABSTRACT

Effective treatment of lung cancer remains a significant clinical challenge due to its multidrug resistance and side effects of the current treatment options. The high mortality associated with this malignancy indicates the need for new therapeutic interventions with fewer side effects. Natural compounds offer various benefits such as easy access, minimal side effects, and multi-molecular targets and thus, can prove useful in treating lung cancer. Sanguinarine (SNG), a natural compound, possesses favorable therapeutic potential against a variety of cancers. Here, we examined the underlying molecular mechanisms of SNG in Non-Small Cell Lung Cancer (NSCLC) cells. SNG suppressed cell growth and induced apoptosis via downregulation of the constitutively active JAK/STAT pathway in all the NSCLC cell lines. siRNA silencing of STAT3 in NSCLC cells further confirmed the involvement of the JAK/STAT signaling cascade. SNG treatment increased Bax/Bcl-2 ratio, which contributed to a leaky mitochondrial membrane leading to cytochrome c release accompanied by caspase activation. In addition, we established the antitumor effects of SNG through reactive oxygen species (ROS) production, as inhibiting ROS production prevented the apoptosis-inducing potential of SNG. In vivo xenograft tumor model further validated our in vitro findings. Overall, our study investigated the molecular mechanisms by which SNG induces apoptosis in NSCLC, providing avenues for developing novel natural compound-based cancer therapies.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Benzophenanthridines/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Isoquinolines/pharmacology , Janus Kinases/drug effects , Lung Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , RNA, Small Interfering/pharmacology , STAT3 Transcription Factor , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
18.
Sci Rep ; 11(1): 17759, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493746

ABSTRACT

Inducing cardiac myocytes to proliferate is considered a potential therapy to target heart disease, however, modulating cardiac myocyte proliferation has proven to be a technical challenge. The Hippo pathway is a kinase signaling cascade that regulates cell proliferation during the growth of the heart. Inhibition of the Hippo pathway increases the activation of the transcription factors YAP/TAZ, which translocate to the nucleus and upregulate transcription of pro-proliferative genes. The Hippo pathway regulates the proliferation of cancer cells, pluripotent stem cells, and epithelial cells through a cell-cell contact-dependent manner, however, it is unclear if cell density-dependent cell proliferation is a consistent feature in cardiac myocytes. Here, we used cultured human iPSC-derived cardiac myocytes (hiCMs) as a model system to investigate this concept. hiCMs have a comparable transcriptome to the immature cardiac myocytes that proliferate during heart development in vivo. Our data indicate that a dense syncytium of hiCMs can regain cell cycle activity and YAP expression and activity when plated sparsely or when density is reduced through wounding. We found that combining two small molecules, XMU-MP-1 and S1P, increased YAP activity and further enhanced proliferation of low-density hiCMs. Importantly, these compounds had no effect on hiCMs within a dense syncytium. These data add to a growing body of literature that link Hippo pathway regulation with cardiac myocyte proliferation and demonstrate that regulation is restricted to cells with reduced contact inhibition.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/physiology , Base Sequence , Cell Count , Cell Cycle/drug effects , Cell Differentiation , Cell Division/drug effects , Cells, Cultured , Contact Inhibition/drug effects , Hippo Signaling Pathway , Humans , Induced Pluripotent Stem Cells/metabolism , Lysophospholipids/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide , RNA/biosynthesis , RNA/genetics , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sulfonamides/pharmacology , Transcription Factors/physiology , YAP-Signaling Proteins
19.
Cells ; 10(8)2021 07 30.
Article in English | MEDLINE | ID: mdl-34440705

ABSTRACT

The aim of the study was to verify the hypothesis that a potential cause of the phytotoxicity of diclofenac (DCF, a non-steroidal anti-inflammatory drug) is an effect of cell cycle progression. This research was conducted using synchronous cultures of a model organism, green alga Chlamydomonas reinhardtii. The project examined DCF effects on selected parameters that characterize cell cycle progression, such as cell size, attainment of commitment points, DNA replication, number of nuclei formed during cells division and morphology of cells in consecutive stages of the cell cycle, together with the physiological and biochemical parameters of algae cells at different stages. We demonstrated that individual cell growth remained unaffected, whereas cell division was delayed in the DCF-treated groups grown in continuous light conditions, and the number of daughter cells from a single cell decreased. Thus, the cell cycle progression is a target affected by DCF, which has a similar anti-proliferative effect on mammalian cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Cell Cycle/drug effects , Cell Division/drug effects , Chlamydomonas reinhardtii/drug effects , Diclofenac/toxicity , Cell Size/drug effects , Chlamydomonas reinhardtii/genetics , Chlamydomonas reinhardtii/growth & development , DNA Replication/drug effects , DNA, Plant/biosynthesis , DNA, Plant/genetics , Photosynthesis/drug effects
20.
Molecules ; 26(16)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34443472

ABSTRACT

Feruloylacetone (FER) is a natural degradant of curcumin after heating, which structurally reserves some functional groups of curcumin. It is not as widely discussed as its original counterpart has been previously; and in this study, its anticancer efficacy is investigated. This study focuses on the suppressive effect of FER on colon cancer, as the efficacious effect of curcumin on this typical cancer type has been well evidenced. In addition, demethoxy-feruloylacetone (DFER) was applied to compare the effect that might be brought on by the structural differences of the methoxy group. It was revealed that both FER and DFER inhibited the proliferation of HCT116 cells, possibly via suppression of the phosphorylated mTOR/STAT3 pathway. Notably, FER could significantly repress both the STAT3 phosphorylation and protein levels. Furthermore, both samples showed capability of arresting HCT116 cells at the G2/M phase via the activation of p53/p21 and the upregulation of cyclin-B. In addition, ROS elevation and changes in mitochondrial membrane potential were revealed, as indicated by p-atm elevation. The apoptotic rate rose to 36.9 and 32.2% after being treated by FER and DFER, respectively. In summary, both compounds exhibited an anticancer effect, and FER showed a greater proapoptotic effect, possibly due to the presence of the methoxy group on the aromatic ring.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Curcumin/pharmacology , Mitochondria/drug effects , Styrenes/pharmacology , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Apoptosis/drug effects , Cell Division/drug effects , Colonic Neoplasms/drug therapy , Curcumin/chemistry , Curcumin/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/agonists , G2 Phase/drug effects , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Phenol/chemistry , Phenol/pharmacology , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Styrenes/chemistry , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...