Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Microb Cell Fact ; 23(1): 82, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38481270

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most commonly diagnosed cancers, posing a serious public health challenge that necessitates the development of new therapeutics, therapies, and prevention methods. Among the various therapeutic approaches, interventions involving lactic acid bacteria (LAB) as probiotics and postbiotics have emerged as promising candidates for treating and preventing CRC. While human-isolated LAB strains are considered highly favorable, those sourced from environmental reservoirs such as dairy and fermented foods are also being recognized as potential sources for future therapeutics. RESULTS: In this study, we present a novel and therapeutically promising strain, Lactococcus lactis ssp. lactis Lc4, isolated from dairy sources. Lc4 demonstrated the ability to release the cytostatic agent - arginine deiminase (ADI) - into the post-cultivation supernatant when cultured under conditions mimicking the human gut environment. Released arginine deiminase was able to significantly reduce the growth of HT-29 and HCT116 cells due to the depletion of arginine, which led to decreased levels of c-Myc, reduced phosphorylation of p70-S6 kinase, and cell cycle arrest. The ADI release and cytostatic properties were strain-dependent, as was evident from comparison to other L. lactis ssp. lactis strains. CONCLUSION: For the first time, we unveil the anti-proliferative properties of the L. lactis cell-free supernatant (CFS), which are independent of bacteriocins or other small molecules. We demonstrate that ADI, derived from a dairy-Generally Recognized As Safe (GRAS) strain of L. lactis, exhibits anti-proliferative activity on cell lines with different levels of argininosuccinate synthetase 1 (ASS1) expression. A unique feature of the Lc4 strain is also its capability to release ADI into the extracellular space. Taken together, we showcase L. lactis ADI and the Lc4 strain as promising, potential therapeutic agents with broad applicability.


Subject(s)
Cytostatic Agents , Lactococcus lactis , Humans , Cytostatic Agents/metabolism , Lactococcus lactis/metabolism , Hydrolases/metabolism , Cell Line, Tumor , Arginine
2.
Nutrients ; 14(19)2022 Sep 24.
Article in English | MEDLINE | ID: mdl-36235622

ABSTRACT

Herein we gathered updated knowledge regarding the alterations of gut microbiota (dysbiosis) and its correlation with human neurodegenerative and brain-related diseases, e.g., Alzheimer's and Parkinson's. This review underlines the importance of gut-derived metabolites and gut metabolic status as the main players in gut-brain crosstalk and their implications on the severity of neural conditions. Scientific evidence indicates that the administration of probiotic bacteria exerts beneficial and protective effects as reduced systemic inflammation, neuroinflammation, and inhibited neurodegeneration. The experimental results performed on animals, but also human clinical trials, show the importance of designing a novel microbiota-based probiotic dietary supplementation with the aim to prevent or ease the symptoms of Alzheimer's and Parkinson's diseases or other forms of dementia or neurodegeneration.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Nervous System Diseases , Parkinson Disease , Probiotics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Dysbiosis/metabolism , Humans , Metabolome , Nervous System Diseases/metabolism , Parkinson Disease/metabolism , Probiotics/therapeutic use
3.
Toxicol Res ; 38(2): 175-186, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35415080

ABSTRACT

The increasing number of depression cases leads to a greater need for new antidepressant treatment development. It is postulated that antidepressants may harm male fertility, but the cellular mechanism is still poorly understood. The role of growth factors and klotho protein in maintaining normal male reproductive function is well documented. Hence, the study aimed to investigate the effect of the antidepressant drug - imipramine (tricyclic AD), and other substances with antidepressant potential (ALS), administered in combination or in combination with LPS (an animal model of depression) on gene expression and protein synthesis of IGF-2 (insulin-like growth factor 2), TGF-ß1 (transforming growth factor ß1), NGF (nerve growth factor), KGF (keratinocyte growth factor) and protein synthesis of VEGF-A (vascular endothelial growth factor A), IGF-IR (insulin-like growth factor receptor 1), EGFR (epidermal growth factor receptor) and klotho in the testis of mice. Mice were injected intraperitoneally with selected ALS and LPS or 10% DMSO (controls) (n = 7/group) once a day for 14 days. Animals were decapitated and testes collected for RNA and protein purification. PCR and western blot methods were employed for the evaluation of growth factors and klotho expression. The results obtained indicated a decreased level of most of the analyzed genes and proteins, except KGF; its expression increased after treatment with MTEP and IMI administrated individually and after NS-398, and IMI in combination with LPS. Our results may suggest that the tested ALS and LPS can contribute to a reduction of male fertility, but NS-398, IMI, and IMI+NS-398 may also act as stimulants after LPS.

4.
Vitam Horm ; 118: 223-246, 2022.
Article in English | MEDLINE | ID: mdl-35180928

ABSTRACT

Klotho gene was originally recognized as a putative aging-suppressor and its prominent age-regulating effects are mostly attributed to the modulation of mineral homeostasis in the kidney. However, recent studies link alterations in hippocampal Klotho expression with cognitive impairment and neurodegenerative diseases. This suggests that hippocampal neurons require Klotho for health and proper functionality. Klotho protects against neuronal dysfunction and regulates several intracellular signaling pathways including oxidative stress response, inflammation, DNA damage, autophagy, endoplasmic reticulum stress response, and multiple types of cell death. Specifically, this chapter covers the current knowledge as to how Klotho protein affects the hippocampal neuronal cells, with special attention paid to underlying molecular mechanisms, and thus influences hippocampal development, hippocampal-dependent cognition, behavior, and motor skills as well as mediates neurodegenerative processes.


Subject(s)
Glucuronidase , Klotho Proteins , Autophagy , Glucuronidase/physiology , Hippocampus/metabolism , Neurons/metabolism
5.
Free Radic Biol Med ; 180: 153-164, 2022 02 20.
Article in English | MEDLINE | ID: mdl-35063649

ABSTRACT

Cancer is one of the leading causes of death in the modern world. Nowadays, most often treatment methods used in clinical oncology are drug therapies applied as monotherapy or combined therapy. Additionally, recent studies focus on developing approaches with the use of a drug in combination with other factors, not only chemical, to improve the probability and magnitude of therapeutic responses and reduce the possibility of chemoresistance. Such a promising factor seems to be an electromagnetic field (EMF) application. Here, we tested the effect of continuous or pulsed EMF on human cancer cells of different origin treated or not with 3-bromopyruvate, a small and powerful alkylating agent with a broad spectrum of anticancer activities. We provide strong evidence suggesting that ELF-EMF potentiates the anti-cancer activity of 3BP in human cancer cells through inhibition of TNFα secretion leading to irreversible p21/p27-dependent G2/M cell cycle arrest and finally cancer cell death. Our findings suggest a novel approach combining pharmacotherapy with ELF-EMF. In conclusion, electromagnetic field seems to be a potential modulator of anti-cancer efficacy of 3BP while combined therapy offers off-target activity. These features contribute to the development of innovative therapeutic strategies for cancer treatment.


Subject(s)
Electromagnetic Fields , Neoplasms , Humans , Neoplasms/drug therapy , Pyruvates/pharmacology
6.
Int J Mol Sci ; 22(21)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34769286

ABSTRACT

The incidence of depression among humans is growing worldwide, and so is the use of antidepressants. However, our fundamental understanding regarding the mechanisms by which these drugs function and their off-target effects against human sexuality remains poorly defined. The present study aimed to determine their differential toxicity on mouse spermatogenic cells and provide mechanistic data of cell-specific response to antidepressant and neuroleptic drug treatment. To directly test reprotoxicity, the spermatogenic cells (GC-1 spg and GC-2 spd cells) were incubated for 48 and 96 h with amitriptyline (hydrochloride) (AMI), escitalopram (ESC), fluoxetine (hydrochloride) (FLU), imipramine (hydrochloride) (IMI), mirtazapine (MIR), olanzapine (OLZ), reboxetine (mesylate) (REB), and venlafaxine (hydrochloride) (VEN), and several cellular and biochemical features were assessed. Obtained results reveal that all investigated substances showed considerable reprotoxic potency leading to micronuclei formation, which, in turn, resulted in upregulation of telomeric binding factor (TRF1/TRF2) protein expression. The TRF-based response was strictly dependent on p53/p21 signaling and was followed by irreversible G2/M cell cycle arrest and finally initiation of apoptotic cell death. In conclusion, our findings suggest that antidepressants promote a telomere-focused DNA damage response in germ cell lines, which broadens the established view of antidepressants' and neuroleptic drugs' toxicity and points to the need for further research in this topic with the use of in vivo models and human samples.


Subject(s)
Antidepressive Agents/toxicity , Antipsychotic Agents/toxicity , G2 Phase Cell Cycle Checkpoints/drug effects , Micronuclei, Chromosome-Defective/chemically induced , Spermatogenesis/drug effects , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Amitriptyline/toxicity , Animals , Cell Line , Escitalopram/toxicity , Fluoxetine/toxicity , Gene Expression Regulation/drug effects , Imipramine/toxicity , Male , Mice , Mirtazapine/toxicity , Models, Biological , Olanzapine/toxicity , Organ Specificity , Reboxetine/toxicity , Reproduction/drug effects , Signal Transduction/drug effects , Time Factors , Venlafaxine Hydrochloride/toxicity
7.
Acta Histochem ; 123(3): 151685, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33556704

ABSTRACT

Insulin-like growth factor (IGF-1) affects almost all cells in the body. Extremely important functions of this growth factor have been demonstrated in the brain and the reproductive system of both, females and males. Also, it is considered as a pro-inflammatory cytokine adjusting tissue homeostasis. However, it seems to play a special role in the male reproductive system and it may be disturbed by the application of antidepressants with different mechanisms of drug action during therapy. To date, the effect of antidepressant-like substances (ALS) on the course of physiological processes in male testicular cells is poorly understood. Therefore, the purpose of the research was to determine the presence, localizationof IGF-1R (insulin-like growth factor 1 ß receptor) and mRNA gene expression of IGF-1R and IGF-1 after administration of 3-[(2-methyl-1,3-tiazol-4-yl)ethynyl]-pyridine (MTEP) and N-[2-(Cyclohexyloxy)-4-nitrophenyl]-methanesulfonamide (NS-398) in the different scheme in the testis of mice. Imipramine was used as a reference drug having a documented interaction with the mGluR5 receptors. The immunohistochemical analyses showed the localization of IGF-1R in Sertoli, Leydig, and germinal cells after all used substances. Differences in receptor localization were observed depending on the drugs applied and the type of analyzed cells. In contrast, there was a significant increase in IGF-1 gene expression after IMI + NS-398 and in IGF-1R after MTEP + NS-398 and IMI + NS-398 administration. It can, therefore, be assumed that the use of a combination of NS-398 with some ALS may run different mechanisms of action and affect the regulation of reproductive function in mouse testis through maintaining homeostasis at the molecular and immunological levels related to IGF.


Subject(s)
Antidepressive Agents/pharmacology , Gene Expression/drug effects , Insulin-Like Growth Factor I/metabolism , Testis/metabolism , Animals , Disease Models, Animal , Insulin-Like Growth Factor I/pharmacology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
8.
Environ Monit Assess ; 193(3): 129, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33587214

ABSTRACT

Samples of leaves, flowers, soil, pollen, bee workers, bee brood, honey, and beeswax were collected to assess the possibility of a transfer of captan, thiacloprid, penthiopyrad, and λ-cyhalothrin from apple trees of Idared variety to honey bee (Apis mellifera) hives. Chemical analyses were performed using the Agilent 7890 Gas Chromatograph equipped with the Micro-cell Electron Capture Detector. It was found that significant amounts of penthiopyrad, the active ingredient of Fontelis 200 SC, were present in leaves, flowers, pollen, bee workers, and beeswax. Simultaneously, captan was present in the brood, worker bees, and honey samples. Significant levels of the captan residues were also detected on the soil surface. In honey samples, captan residue levels exceeded the acceptable standard, reaching 160% of its maximum residue level. However, in no case the amounts of captan, thiacloprid, penthiopyrad, and λ-cyhalothrin ingested with honey by an adult consumer exceeded the level of 0.02% of the acceptable daily intake. Despite the trace amounts of pesticide residues in honey samples collected during the field trial, bee honey consumption can be considered safe. An adult consumer can safely consume about 16 kg of honey.


Subject(s)
Insecticides , Malus , Animals , Bees , Captan/analysis , Environmental Monitoring , Insecta , Insecticides/analysis , Insecticides/toxicity , Neonicotinoids , Nitriles , Pyrazoles , Pyrethrins , Thiazines , Thiophenes
10.
Planta ; 252(2): 23, 2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32676847

ABSTRACT

MAIN CONCLUSION: The changes in the expression of key sugar metabolism enzymes (SPS and SUS), sucrose content and arrangement of chloroplast starch may play a significant role in the cold response in M. giganteus and maize plants. To understand the mechanism of the chilling-response of two closely-related C4 plants, we investigated the changes in the expression of sucrose phosphate synthase (SPS) and sucrose synthase (SUS) as well as changes in their potential products: sucrose, cellulose and starch in the leaves of Miscanthus × giganteus and Zea mays. Low temperature (12-14 °C) increased SPS content in Miscanthus (MG) and chilling-sensitive maize line (Zm-S), but not in chilling-tolerant one (Zm-T). In Zm-S line, chilling also caused the higher intensity of labelling of SPS in the cytoplasm of mesophyll cells, as demonstrated by electron microscopy. SUS labelling was also increased by cold stress only in MG plants what was observed in the secondary wall between mesophyll and bundle sheath cells, as well as in the vacuoles of companion cells. Cold led to a marked increase in total starch grain area in the chloroplasts of Zm-S line. In turn, Fourier transform infrared spectroscopy (FTIR) showed a slight shift in the cellulose band position, which may indicate the formation of more compact cellulose arrangement in Zm-T maize line. In conclusion, this work presents new findings supporting diversified cold-response, not only between two C4 plant species but also within one species of maize.


Subject(s)
Carbohydrate Metabolism , Glucosyltransferases/metabolism , Poaceae/enzymology , Zea mays/enzymology , Cellulose/metabolism , Chloroplasts/metabolism , Cold Temperature , Immunohistochemistry , Plant Leaves/enzymology , Plant Leaves/physiology , Plant Leaves/ultrastructure , Plant Proteins/metabolism , Poaceae/physiology , Poaceae/ultrastructure , Starch/metabolism , Stress, Physiological , Sucrose/metabolism , Zea mays/physiology , Zea mays/ultrastructure
11.
Cells ; 9(5)2020 05 16.
Article in English | MEDLINE | ID: mdl-32429346

ABSTRACT

Neuroinflammation is defined as the activation of the brain's innate immune system in response to an inflammatory challenge and is considered to be a prominent feature of neurodegenerative diseases. The contribution of overactivated neuroglial cells to neuroinflammation and neurodegenerative disorders is well documented, however, the role of hippocampal neurons in the neuroinflammatory process remains fragmentary. In this study, we show for the first time, that klotho acts as a signal transducer between pro-survival and pro-apoptotic crosstalk mediated by ER stress in HT-22 hippocampal neuronal cells during LPS challenge. In control HT-22 cells, LPS treatment results in activation of the IRE1α-p38 MAPK pathway leading to increased secretion of anti-inflammatory IL-10, and thus, providing adaptation mechanism. On the other hand, in klotho-deficient HT-22 cells, LPS induces oxi-nitrosative stress and genomic instability associated with telomere dysfunctions leading to p53/p21-mediated cell cycle arrest and, in consequence, to ER stress, inflammation as well as of apoptotic cell death. Therefore, these results indicate that klotho serves as a part of the cellular defense mechanism engaged in the protection of neuronal cells against LPS-mediated neuroinflammation, emerging issues linked with neurodegenerative disorders.


Subject(s)
Brain/pathology , Glucuronidase/metabolism , Immune System/pathology , Lipopolysaccharides/pharmacology , Animals , Autophagy/drug effects , Cell Line , DNA/metabolism , Endoplasmic Reticulum Stress/drug effects , Gene Silencing , Hippocampus/pathology , Homeostasis/drug effects , Inflammation/immunology , Inflammation/pathology , Klotho Proteins , Mice , Minerals/metabolism , Models, Biological , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction/drug effects , Telomere/metabolism
12.
Cells ; 9(2)2020 01 21.
Article in English | MEDLINE | ID: mdl-31972978

ABSTRACT

Immunosenescence in monocytes has been shown to be associated with several biochemical and functional changes, including development of senescence-associated secretory phenotype (SASP), which may be inhibited by klotho protein. To date, it was believed that SASP activation is associated with accumulating DNA damage. However, some literature data suggest that endoplasmic reticulum and Golgi stress pathways may be involved in SASP development. Thus, the aim of this study was to investigate the role of klotho protein in the regulation of immunosenescence-associated Golgi apparatus and ER stress response induced by bacterial antigens in monocytes. We provide evidence that initiation of immunosenescent-like phenotype in monocytes is accompanied by activation of CREB34L and TFE3 Golgi stress response and ATF6 and IRE1 endoplasmic reticulum stress response, while klotho overexpression prevents these changes. Further, these changes are followed by upregulated secretion of proinflammatory cytokines, which final modification takes place exclusively in the Golgi apparatus. In conclusion, we provide for the first time evidence of klotho involvement in the crosstalk on the line ER-Golgi, which may, in turn, affect activation of SASP. This data may be useful for a novel potential target for therapy in age-related and chronic inflammatory conditions.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cellular Senescence , Endoplasmic Reticulum Stress , Glucuronidase/metabolism , Golgi Apparatus/metabolism , Monocytes/pathology , Anti-Inflammatory Agents/pharmacology , Cellular Senescence/drug effects , Cytokines/metabolism , Endoplasmic Reticulum Stress/drug effects , Golgi Apparatus/drug effects , Humans , Klotho Proteins , Lipopolysaccharides , Monocytes/drug effects
13.
Prog Biophys Mol Biol ; 150: 203-205, 2020 01.
Article in English | MEDLINE | ID: mdl-31381892

ABSTRACT

The novel cancer theory named 'the tissue organization field theory' (TOFT) suggests that carcinogenesis is a process analogous to embryonic development, whereby organs are formed through interactions among different cell types. The suggested 'morphological remodelling' of the epithelium under hypoxia in gut breathing fish (GBF) has many common features with carcinogenesis. It appears that research into the relationship among epidermal growth factor receptor (EGFR), hypoxia inducible factor (HIF) as well as hypoxia and normoxia states in GBF fishes can be crucial in learning about the steering mechanisms of squamous epithelium proliferation, leading to a better understanding of carcinogenesis.


Subject(s)
Carcinogenesis/metabolism , Cell Hypoxia/physiology , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Hypoxia-Inducible Factor 1/metabolism , Neoplasms/metabolism , Animals , Cell Communication , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/cytology , Fishes , Humans , Lung/metabolism , Models, Biological , Signal Transduction , Urinary Bladder/metabolism
14.
Semin Cancer Biol ; 60: 362-364, 2020 02.
Article in English | MEDLINE | ID: mdl-31622661

ABSTRACT

The tissue organization field theory (TOFT) presented completely new, different from the previous one, perspective of research on neoplasm processes. It implicates that secretory neuroepithelial-like cells (NECs), putative chemoreceptors are probably responsible for the control of squamous epithelial cells proliferation in the digestive tract during hypoxia in gut breathing fish (GBF). On the other hand, chemoreceptors dysfunction can lead to uncontrolled proliferation and risk of cancer development in mammals, including humans. The studies on NECs like cells (signal capturing and transduction) may be crucial for understanding the processes of controlling the proliferation of squamous epithelial cells in the digestive tract of GBF fish during hypoxia states. This knowledge can contribute to the explanation of cancer processes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chemoreceptor Cells/metabolism , Disease Susceptibility , Neoplasms/etiology , Neoplasms/metabolism , Animals , Genetic Predisposition to Disease , Humans , Neoplasms/pathology
15.
Apoptosis ; 25(1-2): 57-72, 2020 02.
Article in English | MEDLINE | ID: mdl-31732843

ABSTRACT

In the previous paper of our group, we have demonstrated that one of the crucial factors involved in the crosstalk between autophagy and apoptosis is klotho protein. We have shown that klotho silencing in normal human fibroblasts intensifies lipopolysaccharide (LPS)-induced p-eIF2a-mediated stress of endoplasmic reticulum and thus leads to retardation of prosurvival autophagy and induction of apoptotic cell death. In this study, we have performed a detailed step-by-step analysis of autophagy flux-related genes' expression and endoplasmic reticulum and Golgi stress related pathways in order to determine the exact mechanistic event when autophagy is inhibited in klotho-deficient cells on account of apoptosis initiation. We provide evidence that klotho-silencing in LPS-treated cells results in differential course of ER- and Golgi-mediated stress response. Further, we show that in klotho-deficient cells formation of ULK1 complex is inhibited and thus autophagy initiation is blocked on the account of apoptosis activation, while in the control cells cytoprotective autophagy is activated. Finally, in klotho-deficient cells formation of ULK1 complex is prevented by downregulated expression of Atg13. Thus, this study suggests a novel targeting pathway for efficient elimination of autophagy-deficient cells.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Endoplasmic Reticulum Stress , Fibroblasts/metabolism , Glucuronidase/metabolism , Golgi Apparatus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Proteins/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Fibroblasts/cytology , Glucuronidase/genetics , Golgi Apparatus/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Klotho Proteins , Protein Binding , Signal Transduction
16.
Cell Tissue Res ; 379(3): 613-624, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31705214

ABSTRACT

The European bison is still an animal endangered with extinction, so by learning factors that regulate its reproduction, we can contribute to the survival of this species. On the other hand, autophagy is a dynamic, lisosomal, and evolutionary conserved process which is essential for animal cell survival, homeostasis, and differentiation. This process was demonstrated in many species and in many organs; however, information on the metabolic course of autophagy in the male reproductive system in seasonally reproducing species is lacking. Therefore, in this study, we examined for the first time several autophagy-related factors (mTOR, ULK1, Atg13, PI3K, beclin1, beclin2, Atg14, Atg5, Atg16L, LC3) in testicular and epididymal tissues obtained from adult male individuals of the European bison. We compared the level of gene expression, protein synthesis, and localization of autophagy-related factors between June, September, and December (before, during, and after reproductive activity, respectively). We confirmed that the induction of autophagy was at the highest level in the period after reproductive activity, i.e., in December, when a significant increase in the gene and protein expression was observed for the majority of these factors, probably to ensure cellular protection. However, autophagy was also clearly marked in September, during the intense spermatogenesis, and this may indicate a great demand for autophagy-related proteins required for the normal development of reproductive cells. Obtained results seem to confirm that autophagy pathway, as a consequence of seasonal reproduction, may control the normal course of spermatogenesis in the male European bison.


Subject(s)
Epididymis/cytology , Testis/cytology , Animals , Autophagy/physiology , Bison , Epididymis/metabolism , Male , Seasons , Testis/metabolism
17.
Apoptosis ; 24(9-10): 773-784, 2019 10.
Article in English | MEDLINE | ID: mdl-31278507

ABSTRACT

Depression is a serious medical condition, typically treated by antidepressants. Conventional monotherapy can be effective only in 60-80% of patients, thus modern psychiatry deals with the challenge of new methods development. At the same moment, interactions between antidepressants and the occurrence of potential side effects raise serious concerns, which are even more exacerbated by the lack of relevant data on exact molecular mechanisms. Therefore, the aims of the study were to provide up-to-date information on the relative mechanisms of action of single antidepressants and their combinations. In this study, we evaluated the effect of single and combined antidepressants administration on mouse hippocampal neurons after 48 and 96 h in terms of cellular and biochemical features in vitro. We show for the first time that co-treatment with amitriptyline/imipramine + fluoxetine initiates in cells adaptation mechanisms which allow cells to adjust to stress and finally exerts less toxic events than in cells treated with single antidepressants. Antidepressants treatment induces in neuronal cells oxidative and nitrosative stress, which leads to micronuclei and double-strand DNA brakes formation. At this point, two different mechanistic events are initiated in cells treated with single and combined antidepressants. Single antidepressants (amitriptyline, imipramine or fluoxetine) activate cell cycle arrest resulting in proliferation inhibition. On the other hand, treatment with combined antidepressants (amitriptyline/imipramine + fluoxetine) initiates p16-dependent cell cycle arrest, overexpression of telomere maintenance proteins and finally restoration of proliferation. In conclusion, our findings may pave the way to better understanding of the stress-related effects on neurons associated with mono- and combined therapy with antidepressants.


Subject(s)
Antidepressive Agents , Depression/drug therapy , Neurons/drug effects , Amitriptyline/pharmacology , Amitriptyline/toxicity , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/toxicity , Cell Cycle Checkpoints/drug effects , Fluoxetine/pharmacology , Fluoxetine/toxicity , Hippocampus/cytology , Imipramine/pharmacology , Imipramine/toxicity , Mice
18.
Mol Neurobiol ; 56(10): 6952-6963, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30945158

ABSTRACT

Amitriptyline, antidepressant frequently prescribed for treatment of depressive disorders and several neuropathic and inflammatory diseases, has been shown to cause neurotoxic effects. This effect has been partially linked with increased oxidative stress and apoptosis initiation; however, the exact mechanism is still unknown. Klotho protein due to its neuroprotective characteristics seems to be involved in the amitriptyline-mediated neurotoxicity. In this study, we have evaluated the effect of klotho silencing on mouse hippocampal cells exposed to amitriptyline. We show, for the first time, that klotho silencing intensified in hippocampal neurons amitriptyline-induced imbalance in oxido-nitrosative and mineral homeostasis, genomic instability associated with telomere dysfunction what resulted in p16- and p53/p21-mediated cell cycle arrest and activation of autophagy and apoptotic cell death in consequence. Therefore, these results indicate that klotho serves as a part of the cellular defense mechanism engaged in the protection of neurons against amitriptyline-mediated toxicity.


Subject(s)
Amitriptyline/toxicity , Apoptosis , Autophagy , Glucuronidase/metabolism , Hippocampus/pathology , Neurons/pathology , Telomeric Repeat Binding Protein 2/metabolism , Animals , Cell Line , DNA Damage , Gene Silencing/drug effects , Genomic Instability , Klotho Proteins , Mice , Neurons/drug effects , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , RNA, Small Interfering/metabolism
19.
Acta Histochem ; 121(2): 151-155, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30528348

ABSTRACT

It is suggested that due to the several stomach modifications, Otocinculus affinis (dwarf sucking catfish) possess the ability to breathe air during hypoxia, however, the exact mechanism remains unknown. The aim of this study was detailed analysis of the expression of EGFR in the stomach and diverticulum of the O. affinis at the mRNA and protein levels together with the immunohistochemical localization of EGFR in these organs. The intensity of band fluorescence corresponding to the EGFR gene expression level is significantly higher in the stomach than in the diverticulum. Further, quantitative analysis of EGFR protein abundance also revealed its higher synthesis in the stomach than in the diverticulum and the immunohistochemistry method confirmed these results. As regional localization of respiratory function in gut air-breathing fishes seems to be connected with "morphological remodeling" of the epithelium of their gut, the present research demonstrated the potential efficiency of the O. affinis stomach as a respiratory organ. Having the potential possibility to create an air-blood barrier in the gastrointestinal tract allowing gas diffusion and respiration in hypoxic states seems to be very beneficial for these fish. It seems that facultative gut air breathing fish species are a relevant vertebrate model for high throughput screening, vascular biology and evolution.


Subject(s)
Diverticulum/metabolism , Epithelium/metabolism , ErbB Receptors/metabolism , Gastric Mucosa/metabolism , Adaptation, Physiological/physiology , Animals , Catfishes , Hypoxia/metabolism , Immunohistochemistry/methods , Oxygen Consumption/physiology , Stomach/pathology
20.
Apoptosis ; 24(1-2): 95-107, 2019 02.
Article in English | MEDLINE | ID: mdl-30357572

ABSTRACT

Bacterial endotoxins have been shown to induce prosurvival autophagy or apoptosis in fibroblasts and thus impair the wound healing process. Endoplasmic reticulum has been proposed as a molecular switch between these processes and klotho protein possessing pleiotropic characteristics seems to be involved in both processes, however the exact molecular mechanism is unknown. In this study, we have evaluated the effect of klotho silencing on human fibroblasts exposed to a non-toxic dose of lipopolysaccharide in terms of in vitro wound healing ability. We show for the first time, that klotho silencing in fibroblasts intensified lipopolysaccharide-induced oxidative stress and inflammatory response, what resulted in genomic instability, p-eIF2a-mediated ER stress, retardation of prosurvival autophagy, induction of apoptotic cell death and finally in impaired wound closure. Therefore, our data suggest that klotho serves as a part of cellular defense mechanism engaged in providing protection against bacterial infections during wound healing by modulating ER-signaling crosstalk between autophagy and apoptosis.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum/drug effects , Glucuronidase/physiology , Lipopolysaccharides , Apoptosis/genetics , Autophagy/genetics , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Infections/pathology , Cell Death/drug effects , Cell Death/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Klotho Proteins , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects , Wound Healing/drug effects , Wound Healing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...