Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.042
Filter
1.
Int J Mol Sci ; 23(13)2022 Jun 29.
Article in English | MEDLINE | ID: mdl-35806221

ABSTRACT

There is currently a controversial and heated debate about the safety and ethical aspects of fluoride (F) used for human consumption. Thus, this study assessed the effects of prenatal and postnatal F exposure of rats on the salivary glands of their offspring. Pregnant rats were exposed to 0, 10, or 50 mg F/L from the drinking water, from the first day of gestation until offspring weaning (42 days). The offspring rats were euthanized for the collection of the parotid (PA) and submandibular (SM) glands, to assess the oxidative biochemistry and to perform morphometric and immunohistochemical analyses. F exposure was associated with a decrease in the antioxidant competence of PA in the 10 mg F/L group, contrasting with the increase observed in the 50 mg F/L group. On the other hand, the antioxidant competence of the SM glands was decreased at both concentrations. Moreover, both 10 and 50 mg F/L groups showed lower anti-α-smooth muscle actin immunostaining area in SM, while exposure to 50 mg F/L was associated with changes in gland morphometry by increasing the duct area in both glands. These findings demonstrate a greater susceptibility of the SM glands of the offspring to F at high concentration in comparison to PA, reinforcing the need to adhere to the optimum F levels recommended by the regulatory agencies. Such findings must be interpreted with caution, especially considering their translational meaning.


Subject(s)
Fluorides , Maternal Exposure , Parotid Gland , Submandibular Gland , Animals , Animals, Newborn , Cell Size/drug effects , Female , Fluorides/toxicity , Immunohistochemistry , Keratin-18/metabolism , Lactation , Male , Oxidative Stress/drug effects , Parotid Gland/drug effects , Parotid Gland/metabolism , Parotid Gland/pathology , Pregnancy , Rats , Rats, Wistar , Submandibular Gland/drug effects , Submandibular Gland/metabolism , Submandibular Gland/pathology
2.
Biochem J ; 479(3): 401-424, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35147166

ABSTRACT

The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade promotes cardiomyocyte hypertrophy and is cardioprotective, with the three RAF kinases forming a node for signal integration. Our aims were to determine if BRAF is relevant for human heart failure, whether BRAF promotes cardiomyocyte hypertrophy, and if Type 1 RAF inhibitors developed for cancer (that paradoxically activate ERK1/2 at low concentrations: the 'RAF paradox') may have the same effect. BRAF was up-regulated in heart samples from patients with heart failure compared with normal controls. We assessed the effects of activated BRAF in the heart using mice with tamoxifen-activated Cre for cardiomyocyte-specific knock-in of the activating V600E mutation into the endogenous gene. We used echocardiography to measure cardiac dimensions/function. Cardiomyocyte BRAFV600E induced cardiac hypertrophy within 10 d, resulting in increased ejection fraction and fractional shortening over 6 weeks. This was associated with increased cardiomyocyte size without significant fibrosis, consistent with compensated hypertrophy. The experimental Type 1 RAF inhibitor, SB590885, and/or encorafenib (a RAF inhibitor used clinically) increased ERK1/2 phosphorylation in cardiomyocytes, and promoted hypertrophy, consistent with a 'RAF paradox' effect. Both promoted cardiac hypertrophy in mouse hearts in vivo, with increased cardiomyocyte size and no overt fibrosis. In conclusion, BRAF potentially plays an important role in human failing hearts, activation of BRAF is sufficient to induce hypertrophy, and Type 1 RAF inhibitors promote hypertrophy via the 'RAF paradox'. Cardiac hypertrophy resulting from these interventions was not associated with pathological features, suggesting that Type 1 RAF inhibitors may be useful to boost cardiomyocyte function.


Subject(s)
Cardiomegaly/pathology , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins B-raf/physiology , Animals , Carbamates/pharmacology , Carbamates/toxicity , Cardiomegaly/metabolism , Cell Size/drug effects , Cells, Cultured , Dimerization , Gene Knock-In Techniques , Heart Failure/pathology , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Point Mutation , Protein Conformation/drug effects , Protein Interaction Mapping , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/biosynthesis , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Sulfonamides/toxicity
3.
Toxicol Appl Pharmacol ; 435: 115831, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34922950

ABSTRACT

Nicofluprole is a novel insecticide of the phenylpyrazole class conferring selective antagonistic activity on insect GABA receptors. After repeated daily dietary administration to Wistar rats for 28/90 days, Nicofluprole induced increases in thyroid (and liver) weight, associated with histopathology changes. Nicofluprole did not inhibit thyroid peroxydase nor sodium/iodide symporter, two key players in the biosynthesis of thyroid hormones, indicating the absence of a direct thyroid effect. The results seen in rats suggested a mode of action of Nicofluprole driven by the molecular initiating event of CAR/PXR nuclear receptor activation in livers, with key events of increases in liver weight and hypertrophy, decreasing circulatory thyroid hormones, a compensatory increase in TSH release and follicular cell hypertrophy. To explore the relevance of these changes to humans, well established in vitro rat and human sandwich-cultured hepatocytes were exposed to Nicofluprole up to 7 days. A concentration-dependent CYP3A induction (PXR-activation), an increase in T4-glucuronoconjugation accompanied by UGT1A/2B inductions was observed in rat but not in human hepatocytes. The inductions seen with Nicofluprole in rat (in vivo and in vitro in hepatocytes) that were absent in human hepatocytes represent another example of species-selectivity of nuclear CAR/PXR receptor activators. Importantly, the different pattern observed in rat and human models demonstrate that Nicofluprole-related thyroid effects observed in the rat are with no human relevance.


Subject(s)
Endocrine Disruptors/toxicity , Insecticides/toxicity , Thyroid Gland/drug effects , Animals , Cell Size/drug effects , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Iodide Peroxidase/metabolism , Liver/drug effects , Liver/growth & development , Organ Size/drug effects , Rats , Rats, Wistar , Species Specificity , Symporters/metabolism , Thyroid Gland/pathology , Thyroid Hormones/blood , Thyrotropin/blood
4.
Int J Mol Sci ; 22(24)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34948029

ABSTRACT

Senolytic agents eliminate senescent cells and are expected to reduce senescent cell-mediated adverse effects in cancer therapy. However, the effects of senolytic agents on the survival of irradiated cancer cells remain unknown. Here, the effects of the senolytic agent ABT-263 on the survival of irradiated A549 and Ca9-22 cancer cells were investigated. ABT-263 was added to the culture medium after irradiation. SA-ß-gal activity and cell size, which are hallmarks of cell senescence, were evaluated using a flow cytometer. The colony-forming assay and annexin V staining were performed to test cell survival. We first confirmed that radiation increased the proportion of cells with high SA-ß-gal activity and that ABT-263 decreased it. Of note, ABT-263 decreased the survival of irradiated cancer cells and increased the proportion of radiation-induced annexin V+ cells. Furthermore, the caspase inhibitor suppressed the ABT-263-induced decrease in the survival of irradiated cells. Intriguingly, ABT-263 decreased the proportion of SA-ß-gal low-activity/large cells in the irradiated A549 cells, which was recovered by the caspase inhibitor. Together, these findings suggest that populations maintaining the ability to proliferate existed among the irradiated cancer cells showing senescence-related features and that ABT-263 eliminated the population, which led to decreased survival of irradiated cancer cells.


Subject(s)
Aniline Compounds/pharmacology , Neoplasms/metabolism , Senotherapeutics/pharmacology , Sulfonamides/pharmacology , beta-Galactosidase/metabolism , A549 Cells , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Size/drug effects , Cell Size/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Neoplasms/drug therapy , Neoplasms/radiotherapy , Ultraviolet Rays/adverse effects
5.
Cells ; 10(11)2021 11 16.
Article in English | MEDLINE | ID: mdl-34831419

ABSTRACT

BACKGROUND: To obtain new insights into the activation of the thyroid-stimulating hormone (TSH) and insulin-like growth factor 1 (IGF-1) receptors in human orbital fibroblasts (n-HOFs), the effects of the prostanoid EP2 agonist, omidenepag (OMD), and a rho-associated coiled-coil-containing protein kinase (ROCK) inhibitor, ripasudil (Rip) were evaluated using three-dimension (3D) n-HOFs spheroids in the absence and presence of the recombinant human TSH receptor antibodies, M22 and IGF-1. METHODS: The effects of 100 nM OMD or 10 µM Rip on the physical properties, size, stiffness, and mRNA expression of several extracellular matrix (ECM) molecules, their regulator, inflammatory cytokines, and endoplasmic reticulum (ER) stress-related factors were examined and compared among 3D spheroids of n-HOFs, M22-/IGF-1-activated n-HOFs and GO-related human orbital fibroblasts (GHOFs). RESULTS: The physical properties and mRNA expressions of several genes of the 3D n-HOFs spheroids were significantly and diversely modulated by the presence of OMD or Rip. The OMD-induced effects on M22-/IGF-1-activated n-HOFs were similar to the effects caused by GHOHs, but quite different from those of n-HOFs. CONCLUSIONS: The findings presented herein indicate that the changes induced by OMD may be useful in distinguishing between n-HOFs and GHOFs.


Subject(s)
Fibroblasts/pathology , Glycine/analogs & derivatives , Graves Ophthalmopathy/diagnosis , Graves Ophthalmopathy/pathology , Orbit/pathology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Spheroids, Cellular/pathology , Cell Size/drug effects , Cytokines/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Glycine/pharmacology , Graves Ophthalmopathy/genetics , Humans , Isoquinolines/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Thyrotropin/metabolism , Spheroids, Cellular/drug effects , Sulfonamides/pharmacology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
6.
Cell Rep ; 37(1): 109788, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610309

ABSTRACT

Functional precision medicine aims to match individual cancer patients to optimal treatment through ex vivo drug susceptibility testing on patient-derived cells. However, few functional diagnostic assays have been validated against patient outcomes at scale because of limitations of such assays. Here, we describe a high-throughput assay that detects subtle changes in the mass of individual drug-treated cancer cells as a surrogate biomarker for patient treatment response. To validate this approach, we determined ex vivo response to temozolomide in a retrospective cohort of 69 glioblastoma patient-derived neurosphere models with matched patient survival and genomics. Temozolomide-induced changes in cell mass distributions predict patient overall survival similarly to O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation and may aid in predictions in gliomas with mismatch-repair variants of unknown significance, where MGMT is not predictive. Our findings suggest cell mass is a promising functional biomarker for cancers and drugs that lack genomic biomarkers.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/pathology , Cell Size/drug effects , Glioblastoma/pathology , Single-Cell Analysis/methods , Antineoplastic Agents, Alkylating/therapeutic use , Biomarkers, Tumor/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Drug Screening Assays, Antitumor/methods , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Models, Biological , Neoplasm Grading , Promoter Regions, Genetic , Retrospective Studies , Survival Rate , Temozolomide/pharmacology , Temozolomide/therapeutic use , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
Diabetes ; 70(12): 2810-2822, 2021 12.
Article in English | MEDLINE | ID: mdl-34610982

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce the risk of major adverse cardiovascular (CV) events and hospitalization for heart failure (HF) in patients with type 2 diabetes (T2D). Using CV MRI (CMR) and 31P-MRS in a longitudinal cohort study, we aimed to investigate the effects of the selective SGLT2 inhibitor empagliflozin on myocardial energetics and cellular volume, function, and perfusion. Eighteen patients with T2D underwent CMR and 31P-MRS scans before and after 12 weeks' empagliflozin treatment. Plasma N-terminal prohormone B-type natriuretic peptide (NT-proBNP) levels were measured. Ten volunteers with normal glycemic control underwent an identical scan protocol at a single visit. Empagliflozin treatment was associated with significant improvements in phosphocreatine-to-ATP ratio (1.52 to 1.76, P = 0.009). This was accompanied by a 7% absolute increase in the mean left ventricular ejection fraction (P = 0.001), 3% absolute increase in the mean global longitudinal strain (P = 0.01), 8 mL/m2 absolute reduction in the mean myocardial cell volume (P = 0.04), and 61% relative reduction in the mean NT-proBNP (P = 0.05) from baseline measurements. No significant change in myocardial blood flow or diastolic strain was detected. Empagliflozin thus ameliorates the "cardiac energy-deficient" state, regresses adverse myocardial cellular remodeling, and improves cardiac function, offering therapeutic opportunities to prevent or modulate HF in T2D.


Subject(s)
Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Glucosides/pharmacology , Heart/drug effects , Aged , Benzhydryl Compounds/therapeutic use , Cell Size/drug effects , Cohort Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/prevention & control , Energy Metabolism/drug effects , Female , Glucosides/therapeutic use , Humans , Longitudinal Studies , Male , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , United Kingdom
8.
Biomolecules ; 11(10)2021 10 03.
Article in English | MEDLINE | ID: mdl-34680085

ABSTRACT

In disease states, mesothelial cells are exposed to variable osmotic conditions, with high osmotic stress exerted by peritoneal dialysis (PD) fluids. They contain unphysiologically high concentrations of glucose and result in major peritoneal membrane transformation and PD function loss. The effects of isotonic entry of urea and myo-inositol in hypertonic (380 mOsm/kg) medium on the cell volume of primary cultures of rat peritoneal mesothelial cells and rat kidney outer medullary collecting duct (OMCD) principal cells were studied. In hypertonic medium, rat peritoneal mesothelial cells activated a different mechanism of cell volume regulation in the presence of isotonic urea (100 mM) in comparison to rat kidney OMCD principal cells. In kidney OMCD cells inflow of urea into the shrunken cell results in restoration of cell volume. In the shrunken peritoneal mesothelial cells, isotonic urea inflow caused a small volume increase and activated regulatory volume decrease (RVD). Isotonic myo-inositol activated RVD in hypertonic medium in both cell types. Isotonic application of both osmolytes caused a sharp increase of intracellular calcium both in peritoneal mesothelial cells and in kidney OMCD principal cells. In conclusion, peritoneal mesothelial cells exhibit RVD mechanisms when challenged with myo-inositol and urea under hyperosmolar isotonic switch from mannitol through involvement of calcium-dependent control. Myo-inositol effects were identical with the ones in OMCD principal cells whereas urea effects in OMCD principal cells led to no RVD induction.


Subject(s)
Isotonic Contraction/genetics , Kidney/metabolism , Peritoneal Dialysis , Peritoneum/metabolism , Animals , Cell Size/drug effects , Cells, Cultured , Culture Media/pharmacology , Dialysis Solutions/analysis , Epithelium/chemistry , Epithelium/metabolism , Humans , Inositol/chemistry , Inositol/pharmacology , Osmolar Concentration , Osmotic Pressure/drug effects , Peritoneum/drug effects , Peritoneum/pathology , Primary Cell Culture , Rats
9.
Cells ; 10(10)2021 10 14.
Article in English | MEDLINE | ID: mdl-34685727

ABSTRACT

Multicellular spheroids show three-dimensional (3D) organization with extensive cell-cell and cell-extracellular matrix interactions. Owing to their native tissue-mimicking characteristics, mesenchymal stem cell (MSC) spheroids are considered promising as implantable therapeutics for stem cell therapy. Herein, we aim to further enhance their therapeutic potential by tuning the cultivation parameters and thus the inherent niche of 3D MSC spheroids. Significantly increased expression of multiple pro-regenerative paracrine signaling molecules and immunomodulatory factors by MSCs was observed after optimizing the conditions for spheroid culture. Moreover, these alterations in cellular behaviors may be associated with not only the hypoxic niche developed in the spheroid core but also with the metabolic reconfiguration of MSCs. The present study provides efficient methods for manipulating the therapeutic capacity of 3D MSC spheroids, thus laying solid foundations for future development and clinical application of spheroid-based MSC therapy for regenerative medicine.


Subject(s)
Immunomodulation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Stem Cell Niche , Autophagy/drug effects , Cell Hypoxia/drug effects , Cell Size/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Hydrogels/pharmacology , Immunomodulation/drug effects , Mesenchymal Stem Cells/drug effects , Paracrine Communication/drug effects , Spheroids, Cellular/drug effects , Stem Cell Niche/drug effects
10.
Drug Deliv ; 28(1): 2024-2032, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34569890

ABSTRACT

Cancer, also known as a malignant tumor, has developed into a type of disease with the highest fatality rate, seriously threatening the lives and health of people. Chemotherapy is one of the most important methods for the treatment of cancer. However, chemotherapy drugs have some problems, such as low solubility and lack of targeting, which severely limit their clinical applications. To solve these problems, we designed a block copolymer that has a disulfide bond response. The polymer uses RGD peptide (arginine-glycine-aspartic acid) as the active targeting group, PEG (polyethylene glycol) as the hydrophilic end, and PCL (polycaprolactone) as the hydrophobic end. Then we utilized the amphiphilic polymer as a carrier to simultaneously deliver DOC (docetaxel) and ICG (indocyanine green), to realize the combined application of chemotherapy and photothermal therapy. The antitumor efficacy in vivo and histology analysis showed that the DOC/ICG-loaded micelle exhibited higher antitumor activity. The drug delivery system improved the solubility of DOC and the stability of ICG, realized NIR-guided photothermal therapy, and achieved an ideal therapeutic effect.


Subject(s)
Antineoplastic Agents/administration & dosage , Docetaxel/administration & dosage , Indocyanine Green/administration & dosage , Micelles , Nanoparticles/chemistry , Phototherapy/methods , Animals , Cell Size/drug effects , Chemistry, Pharmaceutical/methods , Docetaxel/pharmacology , Drug Carriers/chemistry , Drug Liberation , Drug Stability , Indocyanine Green/pharmacology , Mice , Mice, Inbred BALB C , Oligopeptides/chemistry , Photothermal Therapy/methods , Polyesters/chemistry , Polyethylene Glycols/chemistry
11.
Mol Biol Rep ; 48(10): 6845-6855, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34476740

ABSTRACT

BACKGROUND: Nuclear factor-κB (NF-κB) has been identified as the major link between inflammation and cancer. Natural agents that inhibit this pathway are essential in attenuating inflammation induced by cancer or chemotherapeutic drugs. High intake of Brassicaceae vegetables has been determined to modulate essential pathways related to chronic diseases. In this study, we investigated the anti-proliferative and anti-inflammatory effects of the indole glucosinolates; indole-3-carbinol (I3C) and its metabolite 3,3-diindolylmethane (DIM) on the inflammatory biomarkers and miRNAs controlling the NF-κB pathway. METHODS AND RESULTS: In our study, we inoculated Ehrlich ascites carcinoma (EAC) cells in female albino mice, which increased their packed cell volume and induced a significant increase in the levels of several cytokines and inflammatory biomarkers (NF-κB IL-6, IL-1b, TNF-α, and NO). A significant elevation in inflammatory-medicated miRNAs (miR-31 and miR-21) was also noted. Treatment with 5-fluorouracil (5-FU) significantly reduced packed cell volume and viable cell count. However, it was accompanied by a significant increase in the levels of inflammatory markers and expression of miR-31 and miR-21. Nevertheless, although treatment with indoles (I3C and DIM) significantly reduced the packed cell volume and viable cell count, their prominent effect was the marked reduction of all inflammatory biomarkers compared to both the EAC untreated group and the EAC group treated with 5-FU. Moreover, the anti-inflammatory effect of I3C or DIM was accompanied by a significant decrease in the expression of miR-31 and miR-21. CONCLUSION: Our findings have; therefore, revealed that I3C and DIM have strong anti-inflammatory effects, implying that their use as a co-treatment with chemotherapeutic drugs can effectively improve the anti-tumor effect of chemotherapeutic drugs.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Ehrlich Tumor/genetics , Glucosinolates/therapeutic use , Indoles/therapeutic use , Inflammation/genetics , MicroRNAs/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers, Tumor/blood , Body Weight/drug effects , Carcinoma, Ehrlich Tumor/blood , Carcinoma, Ehrlich Tumor/pathology , Cell Proliferation/drug effects , Cell Size/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucosinolates/pharmacology , Indoles/pharmacology , Inflammation/blood , Inflammation/pathology , Kidney/drug effects , Kidney/physiopathology , Liver/drug effects , Liver/physiopathology , Mice , MicroRNAs/metabolism , NF-kappa B/metabolism
12.
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
13.
Int J Mol Sci ; 22(13)2021 Jun 24.
Article in English | MEDLINE | ID: mdl-34202704

ABSTRACT

The aim of this study was to evaluate the effect of everolimus, a mammalian target of rapamycin (mTOR) inhibitor, on red blood cell parameters in the context of iron homeostasis in patients with tuberous sclerosis complex (TSC) and evaluate its effect on cell size in vitro. Everolimus has a significant impact on red blood cell parameters in patients with TSC. The most common alteration was microcytosis. The mean MCV value decreased by 9.2%, 12%, and 11.8% after 3, 6, and 12 months of everolimus treatment. The iron level declined during the first 3 months, and human soluble transferrin receptor concentration increased during 6 months of therapy. The size of K562 cells decreased when cultured in the presence of 5 µM everolimus by approximately 8%. The addition of hemin to the cell culture with 5 µM everolimus did not prevent any decrease in cell size. The stage of erythroid maturation did not affect the response to everolimus. Our results showed that the mTOR inhibitor everolimus caused red blood cell microcytosis in vivo and in vitro. This effect is not clearly related to a deficit of iron and erythroid maturation. This observation confirms that mTOR signaling plays a complex role in the control of cell size.


Subject(s)
Cell Size/drug effects , Erythrocytes/drug effects , Erythrocytes/pathology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adolescent , Biomarkers , Cell Differentiation/drug effects , Cell Line , Child , Child, Preschool , Erythrocyte Indices , Erythrocytes/metabolism , Everolimus/administration & dosage , Everolimus/adverse effects , Everolimus/pharmacology , Flow Cytometry , Humans , Iron/metabolism , K562 Cells , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects
14.
Life Sci ; 282: 119794, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34237312

ABSTRACT

AIMS: Engineered conduction tissues (ECTs) fabricated from cardiac progenitor cells (CPCs) and collagen sponges were precisely targeted for the treatment of atrioventricular conduction block in our previous studies. However, obvious shrinkage and deformation of ECTs was observed during in vitro culture. According to the literature, it can be speculated that basic fibroblast growth factor (bFGF) may downregulate alpha-smooth muscle actin (α-SMA) produced by CPCs to prevent the shrinkage of CPC-engineered conduction tissues. MAIN METHODS: In this study, culture media with or without bFGF were used for both cell culture and 3D tissue construction. The expression of α-SMA and the size change of engineered tissue were analyzed to evaluate the feasibility of adding bFGF to regulate α-SMA expression and shrinkage of constructs. In addition, cardiac-specific examinations were performed to evaluate the effect of bFGF on cardiac tissue formation. KEY FINDINGS: Supplementation with bFGF efficiently relieved shrinkage of engineered tissue by downregulating the expression of α-SMA at both the cellular and 3D tissue levels. Moreover, bFGF had a positive influence on cardiac tissue formation in terms of cell viability, tissue organization and electrical conduction velocity. SIGNIFICANCE: This study provides a guide for both shape control and quality improvement of CPC-engineered cardiac tissues.


Subject(s)
Actins/genetics , Culture Media/pharmacology , Fibroblast Growth Factor 2/pharmacology , Myocardium/cytology , Stem Cells/cytology , Tissue Engineering/methods , Animals , Cell Size/drug effects , Cells, Cultured , Down-Regulation/drug effects , Female , Rats, Sprague-Dawley , Stem Cells/drug effects , Stem Cells/metabolism , Tissue Scaffolds/chemistry
15.
J Biochem Mol Toxicol ; 35(10): e22864, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34309121

ABSTRACT

The retrograde signaling pathway is well conserved from yeast to humans, which regulates cell adaptation during stress conditions and prevents cell death. One of its components, RTG1 encoded Rtg1p in association with Rtg3p communicates between mitochondria, nucleus, and peroxisome during stress for adaptation, by regulation of transcription. The F-box motif protein encoded by YDR131C  constitutes a part of SCF Ydr131c -E3 ligase complex, with unknown function; however, it is known that retrograde signaling is modulated by the E3 ligase complex. This study reports epistasis interaction between YDR131C and RTG1, which regulates cell growth, response to genotoxic stress, decreased apoptosis, resistance to petite mutation, and cell wall integrity. The cells of ydr131cΔrtg1Δ genetic background exhibits growth rate improvement however, sensitivity to hydroxyurea, itraconazole antifungal agent and synthetic indoloquinazoline-based alkaloid (8-fluorotryptanthrin, RK64), which disrupts the cell wall integrity in Saccharomyces cerevisiae. The epistatic interaction between YDR131C and RTG1 indicates a link between protein degradation and retrograde signaling pathways.


Subject(s)
Apoptosis/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , DNA Damage/genetics , Epistasis, Genetic , F-Box Motifs/genetics , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Signal Transduction/genetics , Acetic Acid/pharmacology , Antifungal Agents/pharmacology , Apoptosis/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Enlargement/drug effects , Cell Size/drug effects , DNA Damage/drug effects , Ethidium/pharmacology , Gene Deletion , Hydrogen Peroxide/pharmacology , Hydroxyurea/pharmacology , Itraconazole/pharmacology , Microorganisms, Genetically-Modified , Mutation/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sulfinic Acids/pharmacology
16.
Cell Prolif ; 54(7): e13073, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34096124

ABSTRACT

OBJECTIVES: The treatment of ameloblastoma, an odontogenic epithelial tumour destroying jawbone, mainly depends on radical destructive resections. Other therapeutic options are limited by the characteristics of ameloblastoma, such as high recurrence rates and resistance to radiation and chemotherapy, which implies possible existence of cancer stem cells (CSCs) in ameloblastoma. Here, we identified a putative CSC population in immortalized and primary human ameloblastoma cells and examined possible therapeutic reagents to reduce the CSC population. METHODS: We investigated subpopulations of AM-1 cell line and human ameloblastoma cells using immunocytochemistry and flow cytometry and the effects of Wnt signalling activators on the 2- and 3-dimensional cultured ameloblastoma cells using molecular biological analyses. RESULT: Among heterogenous ameloblastoma cells, small-sized and round-shaped cells were found to be proliferative and expressed a marker of dental epithelial stem cells, SRY-box 2 (Sox2). Exogenous activation of Wnt signalling using glycogen synthase kinase 3ß inhibitors, lithium chloride (LiCl) and valproic acid (VPA), increased the cell size and decreased proliferation of cells and expression of Sox2 in 2 dimensionally cultured AM-1 and human primary ameloblastoma cells. Furthermore, the growth of 3 dimensionally cultured AM-1 cells as suspended or embedded in gel was suppressed by treatment with Wnt signalling activators, VPA and CHIR99021, or antibodies to sclerostin, an antagonist of Wnt signalling. CONCLUSION: We suggest that Wnt signalling activators are potential drug candidates to suppress CSCs in ameloblastoma.


Subject(s)
Lithium Chloride/pharmacology , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway/drug effects , Ameloblastoma/metabolism , Ameloblastoma/pathology , Animals , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Size/drug effects , Down-Regulation/drug effects , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/cytology , SOXB1 Transcription Factors/metabolism , Valproic Acid/pharmacology , beta Catenin/metabolism
17.
Sci Rep ; 11(1): 12763, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140545

ABSTRACT

Epithelial-mesenchymal transition (EMT) in primary tumor cells is a key prerequisite for metastasis initiation. Statins, cholesterol-lowering drugs, can delay metastasis formation in vivo and attenuate the growth and proliferation of tumor cells in vitro. The latter effect is stronger in tumor cells with a mesenchymal-like phenotype than in those with an epithelial one. However, the effect of statins on epithelial cancer cells treated with EMT-inducing growth factors such as transforming growth factor-ß (TGF-ß) remains unclear. Here, we examined the effect of atorvastatin on two epithelial cancer cell lines following TGF-ß treatment. Atorvastatin-induced growth inhibition was stronger in TGF-ß-treated cells than in cells not thusly treated. Moreover, treatment of cells with atorvastatin prior to TGF-ß treatment enhanced this effect, which was further potentiated by the simultaneous reduction in the expression of the statin target enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). Dual pharmacological targeting of HMGCR can thus strongly inhibit the growth and proliferation of epithelial cancer cells treated with TGF-ß and may also improve statin therapy-mediated attenuation of metastasis formation in vivo.


Subject(s)
Atorvastatin/pharmacology , Hydroxymethylglutaryl CoA Reductases/metabolism , Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Biomarkers, Tumor/metabolism , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Size/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Methods Mol Biol ; 2326: 155-165, 2021.
Article in English | MEDLINE | ID: mdl-34097267

ABSTRACT

This chapter describes, in detail, the operational principles and experimental design to analyze the premature death of human red blood cells (RBCs; erythrocytes). Necrosis (i.e., hemolysis), eryptosis, and necroptosis are the three types of cell death thus far known to exist in RBCs, and distinctive markers of each are well established. Here, methods based on flow cytometry are presented in an easily reproducible form. Moreover, manipulation of incubation medium to promote or inhibit certain physiological phenomena, along with a step-by-step approach to examine membrane scrambling, cell volume, surface complexity, calcium activity, oxidative stress, and signal transduction pathways are also discussed.


Subject(s)
Eryptosis , Erythrocytes/cytology , Flow Cytometry/methods , Hemolysis , Necroptosis , Calcium Signaling/drug effects , Cell Size/drug effects , Eryptosis/drug effects , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/pathology , Hemolysis/drug effects , Humans , Necroptosis/drug effects , Oxidative Stress/drug effects , Toxicity Tests/methods
19.
Nutrients ; 13(5)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922631

ABSTRACT

Glycerol monocaprylate (GMC) is a glycerol derivative of medium-chain fatty acids (MCFAs) and is widely used as a preservative in food processing. However, GMC and its hydrolytic acid (octylic acid) have antibacterial properties that may affect the physiology and intestinal microecology of the human body. Therefore, in this study, the effects of two different dosages of GMC (150 and 1600 mg kg-1) on glucose, lipid metabolism, inflammation, and intestinal microecology of normal diet-fed C57BL/6 mice were comprehensively investigated. The obtained results showed that the level of triglycerides (TGs) in the low-dose group down-regulated significantly, and the anti-inflammatory cytokine interleukin 10 (IL-10) significantly increased, while the pro-inflammatory cytokines monocyte chemotactic protein 1 (MCP-1) and interleukin 1beta (IL-1ß) in the high-dose group were significantly decreased. Importantly, GMC promoted the α-diversity of gut microbiota in normal-diet-fed mice, regardless of dosages. Additionally, it was found that the low-dose treatment of GMC significantly increased the abundance of Lactobacillus, while the high-dose treatment of GMC significantly increased the abundance of SCFA-producers such as Clostridiales, Lachnospiraceae, and Ruminococcus. Moreover, the content of short-chain fatty acids (SCFAs) was significantly increased by GMC supplementation. Thus, our research provides a novel insight into the effects of GMC on gut microbiota and physiological characteristics.


Subject(s)
Fatty Acids, Volatile/biosynthesis , Gastrointestinal Microbiome/drug effects , Glycerol/pharmacology , Inflammation/microbiology , Metabolism/drug effects , Adipocytes/drug effects , Animals , Body Weight/drug effects , Cell Size/drug effects , Cytokines/blood , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Glucose/metabolism , Hormones/blood , Inflammation/blood , Inflammation/genetics , Inflammation/pathology , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , Male , Mice, Inbred C57BL
20.
Biochem Biophys Res Commun ; 555: 95-101, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33813282

ABSTRACT

Oral squamous cell carcinoma (OSCC) is a highly aggressive carcinoma with a high incidence of recurrence and distant metastasis. However, the mechanism of epithelial to mesenchymal transition (EMT) during tumor progression and metastasis in OSCC has not yet been fully elucidated. It is well known that the Cl- channel controls cell volume and activates several signaling pathways for cell differentiation. The aim of the present study was to investigate the role of the Cl- channel on EMT in the OSC 20 cell line, which is an OSCC line. OSC-20 cells were cultured with low serum medium containing a Cl- channel blocker NPPB. Morphological changes, gene expression, immunoreactivity, cell volume, and signaling pathway of the NPPB-treated OSC-20 cells were evaluated. The NPPB-treated OSC-20 cells showed typical morphology of mesenchymal cells. The expression levels of the epithelial marker E-cadherin in the NPPB-treated OSC-20 cells were lower than those of the untreated and TGF-ß1-treated OSC-20 cells. On the other hand, mesenchymal markers such as vimentin, ZEB1, and Snail, in the NPPB-treated OSC-20 cells were higher than those in the untreated and TGF-ß1-treated OSC-20 cells. Furthermore, a large number of vimentin-positive cells also appeared in the NPPB-treated OSC-20 cells. Additionally, the cell volume of these cells was significantly increased compared to that of the untreated and TGF-ß1-treated cells. Interestingly, NPPB did not activate the TGF-ß/smad signaling pathway, but activated the Wnt/ß-catenin signaling pathway. These results suggest that Cl- channel dysfunction promoted EMT via activation of the Wnt/ß-catenin signaling pathway in OSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Chloride Channels/metabolism , Epithelial-Mesenchymal Transition , Mouth Neoplasms/pathology , Anthracenes/pharmacology , Antigens, CD/metabolism , Cadherins/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Size/drug effects , Chloride Channels/antagonists & inhibitors , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Humans , Mouth Neoplasms/metabolism , Nitrobenzoates/pharmacology , Trypsin/pharmacology , Wnt Signaling Pathway/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...