Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 2445: 139-169, 2022.
Article in English | MEDLINE | ID: mdl-34972991

ABSTRACT

Anticancer therapy is complicated by the ability of malignant cells to activate cytoprotective autophagy that rescues treated cells. This protocol describes methods for analysis of autophagic process in apoptosis-resistant tumor cells treated with damaging agents. Induction of autophagy in these cells can activate apoptotic death. Protocol provides methods for Western blotting, immunofluorescent analysis, and transfection of cells with fluorescent protein-tagged LC3-encoding plasmids to analyze autophagy. Different approaches to change autophagy in tumor cells are suggested. A special approach is connected with induction of cellular senescence. Senescent cells, which are resistant to apoptosis, are vulnerable to certain damaging agents, in particular, to kinase inhibitors. Methods to induce and analyze senescence are considered. They include detection of proliferation arrest by different ways, mTORC1 activity assay and fluorescent analysis of mTORC1 and lysosome localization as a novel senescence hallmark. Incapability of senescent cells to complete autophagy after damage allows to force them to apoptosis. To demonstrate apoptotic cell death, analysis of caspase activity, Annexin V-FITC binding, DNA fragmentation, and mitochondria and lysosome damage are suggested. The methods described can be applied in studies aimed on developing different strategies of tumor cell elimination through changing autophagy.


Subject(s)
Apoptosis , Autophagy , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cellular Senescence , Mechanistic Target of Rapamycin Complex 1/metabolism
2.
PLoS Biol ; 18(7): e3000564, 2020 07.
Article in English | MEDLINE | ID: mdl-32701952

ABSTRACT

Amyloids are protein aggregates with a highly ordered spatial structure giving them unique physicochemical properties. Different amyloids not only participate in the development of numerous incurable diseases but control vital functions in archaea, bacteria and eukarya. Plants are a poorly studied systematic group in the field of amyloid biology. Amyloid properties have not yet been demonstrated for plant proteins under native conditions in vivo. Here we show that seeds of garden pea Pisum sativum L. contain amyloid-like aggregates of storage proteins, the most abundant one, 7S globulin Vicilin, forms bona fide amyloids in vivo and in vitro. Full-length Vicilin contains 2 evolutionary conserved ß-barrel domains, Cupin-1.1 and Cupin-1.2, that self-assemble in vitro into amyloid fibrils with similar physicochemical properties. However, Cupin-1.2 fibrils unlike Cupin-1.1 can seed Vicilin fibrillation. In vivo, Vicilin forms amyloids in the cotyledon cells that bind amyloid-specific dyes and possess resistance to detergents and proteases. The Vicilin amyloid accumulation increases during seed maturation and wanes at germination. Amyloids of Vicilin resist digestion by gastrointestinal enzymes, persist in canned peas, and exhibit toxicity for yeast and mammalian cells. Our finding for the first time reveals involvement of amyloid formation in the accumulation of storage proteins in plant seeds.


Subject(s)
Amyloid/metabolism , Pisum sativum/metabolism , Seed Storage Proteins/metabolism , Seeds/metabolism , Amyloid/ultrastructure , Detergents/pharmacology , Escherichia coli/metabolism , Ions , Pancreatin/metabolism , Pisum sativum/drug effects , Pepsin A/metabolism , Protein Aggregates , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Saccharomyces cerevisiae/metabolism , Seed Storage Proteins/chemistry , Seed Storage Proteins/pharmacology , Seed Storage Proteins/ultrastructure
3.
Aging (Albany NY) ; 10(11): 3574-3589, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30482887

ABSTRACT

Autophagy is conservative catabolic process that degrades organelles, in particular, mitochondria, and misfolded proteins within the lysosomes, thus maintaining cellular viability. Despite the close relationship between mitochondrial dysfunction and cellular senescence, it is unclear how mitochondria damage can induce autophagy in senescent cells. We show that MEK/ERK suppression induces mitochondria damage followed by apoptosis of senescent Ras-expressing cells. To understand the role of persistent mTORC1 signaling in breaking the cAMPK-induced autophagy caused by mitochondrial damage, we inhibited mTORС1 with low concentrations of pp242. mTORC1 suppression neither restores the AMPK-induced autophagy nor decreases the level of apoptosis upon MEK/ERK inhibition. We discovered the existence of an alternative autophagy-like way that partially increases the viability of senescent cells under suppressed mTORC1. The pp242-treated cells survive due to formation of the non-autophagous LC3-negative vacuoles, which contain the damaged mitochondria and lysosomes with the following excretion the content from the cell. MEK/ERK activity is required to implement this process in senescent cells. Senescent cells exhibit distinctive spatial distribution of organelles and proteins that provides uncoupling of final participants of autophagy. We show that this feature stops the process of cytoprotective autophagy in response to MEK/ERK suppression, thus allowing selective elimination of senescent Ras-expressing cells.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Cellular Senescence/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Animals , Cell Survival , Cellular Senescence/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts , Gene Expression Regulation/drug effects , Genes, ras , Humans , Indoles/pharmacology , MAP Kinase Kinase Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Purines/pharmacology , Rats
4.
Aging (Albany NY) ; 9(11): 2352-2375, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29140794

ABSTRACT

The Ras-Raf-MEK-ERK pathway plays a central role in tumorigenesis and is a target for anticancer therapy. The successful strategy based on the activation of cell death in Ras-expressing cells is associated with the suppression of kinases involved in Ras pathway. However, activation of cytoprotective autophagy overcomes antiproliferative effect of the inhibitors and develops drug resistance. We studied whether cellular senescence induced by HDAC inhibitor sodium butyrate in E1a+cHa-Ras-transformed rat embryo fibroblasts (ERas) and A549 human Ki-Ras mutated lung adenocarcinoma cells would enhance the tumor suppressor effect of MEK/ERK inhibition. Treatment of control ERas cells with PD0325901 for 24 h results in mitochondria damage and apoptotic death of a part of cellular population. However, the activation of AMPK-dependent autophagy overcomes pro-apoptotic effects of MEK/ERK inhibitor and results in restoration of the mitochondria and rescue of viability. Senescent ERas cells do not develop cytoprotective autophagy upon inhibition of MEK/ERK pathway due to spatial dissociation of lysosomes and autophagosomes in the senescent cells. Senescent cells are unable to form the autophagolysosomes and to remove the damaged mitochondria resulting in apoptotic death. Our data show that suppression of MEK/ERK pathway in senescent cells provides a new strategy for elimination of Ras-expressing cells.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cellular Senescence/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Histone Deacetylase Inhibitors/pharmacology , Lung Neoplasms/drug therapy , MAP Kinase Kinase Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , A549 Cells , AMP-Activated Protein Kinases/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Signal Transduction/drug effects , Time Factors
6.
Oncotarget ; 7(21): 31563-71, 2016 May 24.
Article in English | MEDLINE | ID: mdl-26883196

ABSTRACT

Cells undergoing oncogenic transformation frequently inactivate tumor suppressor pathways that could prevent their uncontrolled growth. Among those pathways p53 and p38MAPK pathways play a critical role in regulation of cell cycle, senescence and cell death in response to activation of oncogenes, stress and DNA damage. Consequently, these two pathways are important in determining the sensitivity of tumor cells to anti-cancer treatment. Wild type p53-induced phosphatase, Wip1, is involved in governance of both pathways. Recently, strategies directed to manipulation with Wip1 activity proposed to advance current day anticancer treatment and novel chemical compounds synthesized to improve specificity of manipulation with Wip1 activity. Here we reviewed the history of Wip1 studies in vitro and in vivo, in genetically modified animal models that support Wip1 role in tumorigenesis through regulation of p53 and p38MAPK pathways. Based on our knowledge we propose several recommendations for future more accurate studies of Wip1 interactions with other pathways involved in tumorigenesis using recently developed tools and for adoption of Wip1 manipulation strategies in anti-cancer therapy.


Subject(s)
Protein Phosphatase 2C/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Cycle/genetics , Cell Transformation, Neoplastic/genetics , DNA Damage , Humans , Mutation , Protein Phosphatase 2C/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...