Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Aging Cell ; : e14246, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38895933

ABSTRACT

The transcription factor HSF-1 (heat shock factor 1) acts as a master regulator of heat shock response in eukaryotic cells to maintain cellular proteostasis. The protein has a protective role in preventing cells from undergoing ageing, and neurodegeneration, and also mediates tumorigenesis. Thus, modulating HSF-1 activity in humans has a promising therapeutic potential for treating these pathologies. Loss of HSF-1 function is usually associated with impaired stress tolerance. Contrary to this conventional knowledge, we show here that inactivation of HSF-1 in the nematode Caenorhabditis elegans results in increased thermotolerance at young adult stages, whereas HSF-1 deficiency in animals passing early adult stages indeed leads to decreased thermotolerance, as compared to wild-type. Furthermore, a gene expression analysis supports that in young adults, distinct cellular stress response and immunity-related signaling pathways become induced upon HSF-1 deficiency. We also demonstrate that increased tolerance to proteotoxic stress in HSF-1-depleted young worms requires the activity of the unfolded protein response of the endoplasmic reticulum and the SKN-1/Nrf2-mediated oxidative stress response pathway, as well as an innate immunity-related pathway, suggesting a mutual compensatory interaction between HSF-1 and these conserved stress response systems. A similar compensatory molecular network is likely to also operate in higher animal taxa, raising the possibility of an unexpected outcome when HSF-1 activity is manipulated in humans.

2.
Int J Mol Sci ; 24(19)2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37834309

ABSTRACT

N6-methyladenine (6mA) in the DNA is a conserved epigenetic mark with various cellular, physiological and developmental functions. Although the presence of 6mA was discovered a few years ago in the nuclear genome of distantly related animal taxa and just recently in mammalian mitochondrial DNA (mtDNA), accumulating evidence at present seriously questions the presence of N6-adenine methylation in these genetic systems, attributing it to methodological errors. In this paper, we present a reliable, PCR-based method to determine accurately the relative 6mA levels in the mtDNA of Caenorhabditis elegans, Drosophila melanogaster and dogs, and show that these levels gradually increase with age. Furthermore, daf-2(-)-mutant worms, which are defective for insulin/IGF-1 (insulin-like growth factor) signaling and live twice as long as the wild type, display a half rate at which 6mA progressively accumulates in the mtDNA as compared to normal values. Together, these results suggest a fundamental role for mtDNA N6-adenine methylation in aging and reveal an efficient diagnostic technique to determine age using DNA.


Subject(s)
DNA Methylation , DNA, Mitochondrial , Animals , Dogs , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Adenine/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Aging/genetics , Mammals/metabolism
3.
Nat Aging ; 3(9): 1048-1050, 2023 09.
Article in English | MEDLINE | ID: mdl-37620583

Subject(s)
Longevity , Lysosomes
4.
Int J Mol Sci ; 24(9)2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37175806

ABSTRACT

Autophagy (cellular self-degradation) plays a major role in maintaining the functional integrity (homeostasis) of essentially all eukaryotic cells. During the process, superfluous and damaged cellular constituents are delivered into the lysosomal compartment for enzymatic degradation. In humans, age-related defects in autophagy have been linked to the incidence of various age-associated degenerative pathologies (e.g., cancer, neurodegenerative diseases, diabetes, tissue atrophy and fibrosis, and immune deficiency) and accelerated ageing. Muscle mass decreases at detectable levels already in middle-aged patients, and this change can increase up to 30-50% at age 80. AUTEN-67 and -99, two small-molecule enhancers of autophagy with cytoprotective and anti-ageing effects have been previously identified and initially characterized. These compounds can increase the life span in wild-type and neurodegenerative model strains of the fruit fly Drosophila melanogaster. Adult flies were treated with these AUTEN molecules via feeding. Fluorescence and electron microscopy and Western blotting were used to assess the level of autophagy and cellular senescence. Flying tests were used to measure the locomotor ability of the treated animals at different ages. In the current study, the effects of AUTEN-67 and -99 were observed on striated muscle cells using the Drosophila indirect flight muscle (IFM) as a model. The two molecules were capable of inducing autophagy in IFM cells, thereby lowering the accumulation of protein aggregates and damaged mitochondria, both characterizing muscle ageing. Furthermore, the two molecules significantly improved the flying ability of treated animals. AUTEN-67 and -99 decrease the rate at which striated muscle cells age. These results may have a significant medical relevance that could be further examined in mammalian models.


Subject(s)
Drosophila , Muscle, Striated , Animals , Humans , Middle Aged , Aged, 80 and over , Drosophila melanogaster , Aging , Autophagy , Mammals
5.
Biol Futur ; 73(4): 375-384, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35731422

ABSTRACT

Autophagy is a highly conserved self-degradation process of eukaryotic cells which is required for the effective elimination of damaged and unnecessary cytosolic constituents. Defects in the process can cause the intracellular accumulation of such damages, thereby leading to the senescence and subsequent loss of the affected cell. Defective autophagy hence is implicated in the development of various degenerative processes, including cancer, neurodegenerative diseases, diabetes, tissue atrophy and fibrosis, and immune deficiency, as well as in accelerated aging. The autophagic process is mediated by numerous autophagy-related (ATG) proteins, among which the ATG8/LC3/GABARAP (Microtubule-associated protein 1A/1B-light chain 3/Gammaaminobutyric acid receptor-associated protein) superfamily has a pivotal role in the formation and maturation of autophagosome, a key (macro) autophagic structure (the autophagosome sequesters parts of the cytoplasm which are destined for breakdown). While in the unicellular yeast there is only a single ATG8 protein, metazoan systems usually contain more ATG8 paralogs. ATG8 paralogs generally display tissue-specific expression patterns and their functions are not strictly restricted to autophagy. For example, GABARAP proteins also play a role in intracellular vesicle transport, and, in addition to autophagosome formation, ATG8 also functions in selective autophagy. In this review, we summarize the functional diversity of ATG8/LC3/GABARAP proteins, using tractable genetic models applied in autophagy research.


Subject(s)
Autophagy-Related Protein 8 Family , Autophagy , Evolution, Molecular , Animals , Autophagy/genetics , Autophagy-Related Protein 8 Family/classification , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Protein 8 Family/metabolism , Biological Transport , Transport Vesicles/metabolism , Phylogeny
6.
Int J Mol Sci ; 20(22)2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31752429

ABSTRACT

HSF1 (heat shock factor 1) is an evolutionarily conserved master transcriptional regulator of the heat shock response (HSR) in eukaryotic cells. In response to high temperatures, HSF1 upregulates genes encoding molecular chaperones, also called heat shock proteins, which assist the refolding or degradation of damaged intracellular proteins. Accumulating evidence reveals however that HSF1 participates in several other physiological and pathological processes such as differentiation, immune response, and multidrug resistance, as well as in ageing, neurodegenerative demise, and cancer. To address how HSF1 controls these processes one should systematically analyze its target genes. Here we present a novel database called HSF1Base (hsf1base.org) that contains a nearly comprehensive list of HSF1 target genes identified so far. The list was obtained by manually curating publications on individual HSF1 targets and analyzing relevant high throughput transcriptomic and chromatin immunoprecipitation data derived from the literature and the Yeastract database. To support the biological relevance of HSF1 targets identified by high throughput methods, we performed an enrichment analysis of (potential) HSF1 targets across different tissues/cell types and organisms. We found that general HSF1 functions (targets are expressed in all tissues/cell types) are mostly related to cellular proteostasis. Furthermore, HSF1 targets that are conserved across various animal taxa operate mostly in cellular stress pathways (e.g., autophagy), chromatin remodeling, ribosome biogenesis, and ageing. Together, these data highlight diverse roles for HSF1, expanding far beyond the HSR.


Subject(s)
Heat-Shock Proteins/genetics , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Chromatin Immunoprecipitation/methods , Humans , Mice , Molecular Chaperones/genetics , Proteostasis/genetics , Transcription Factors/genetics
7.
FASEB J ; 33(2): 2372-2387, 2019 02.
Article in English | MEDLINE | ID: mdl-30277819

ABSTRACT

NF-E2-related factor 2 (NRF2) transcription factor has a fundamental role in cell homeostasis maintenance as one of the master regulators of oxidative and electrophilic stress responses. Previous studies have shown that a regulatory connection exists between NRF2 and autophagy during reactive oxygen species-generated oxidative stress. The aim of the present study was to investigate how autophagy is turned off during prolonged oxidative stress, to avoid overeating and destruction of essential cellular components. AMPK is a key cellular energy sensor highly conserved in eukaryotic organisms, and it has an essential role in autophagy activation at various stress events. Here the role of human AMPK and its Caenorhabditis elegans counterpart AAK-2 was explored upon oxidative stress. We investigated the regulatory connection between NRF2 and AMPK during oxidative stress induced by tert-butyl hydroperoxide (TBHP) in HEK293T cells and C. elegans. Putative conserved NRF2/protein skinhead-1 binding sites were found in AMPK/aak-2 genes by in silico analysis and were later confirmed experimentally by using EMSA. After addition of TBHP, NRF2 and AMPK showed a quick activation; AMPK was later down-regulated, however, while NRF2 level remained high. Autophagosome formation and Unc-51-like autophagy activating kinase 1 phosphorylation were initially stimulated, but they returned to basal values after 4 h of TBHP treatment. The silencing of NRF2 resulted in a constant activation of AMPK leading to hyperactivation of autophagy during oxidative stress. We observed the same effects in C. elegans demonstrating the conservation of this self-defense mechanism to save cells from hyperactivated autophagy upon prolonged oxidative stress. We conclude that NRF2 negatively regulates autophagy through delayed down-regulation of the expression of AMPK upon prolonged oxidative stress. This regulatory connection between NRF2 and AMPK may have an important role in understanding how autophagy is regulated in chronic human morbidities characterized by oxidative stress, such as neurodegenerative diseases, certain cancer types, and in metabolic diseases.-Kosztelnik, M., Kurucz, A., Papp, D., Jones, E., Sigmond, T., Barna, J., Traka, M. H., Lorincz, T., Szarka, A., Banhegyi, G., Vellai, T., Korcsmaros, T., Kapuy, O. Suppression of AMPK/aak-2 by NRF2/SKN-1 down-regulates autophagy during prolonged oxidative stress.


Subject(s)
Autophagy , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Protein Kinases/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , DNA-Binding Proteins/genetics , Down-Regulation , HEK293 Cells , Humans , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics
8.
J Cell Sci ; 124(Pt 9): 1510-8, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21502138

ABSTRACT

Autophagy is a lysosome-mediated self-degradation process of eukaryotic cells that, depending on the cellular milieu, can either promote survival or act as an alternative mechanism of programmed cell death (PCD) in terminally differentiated cells. Despite the important developmental and medical implications of autophagy and the main form of PCD, apoptosis, orchestration of their regulation remains poorly understood. Here, we show in the nematode Caenorhabditis elegans, that various genetic and pharmacological interventions causing embryonic lethality trigger a massive cell death response that has both autophagic and apoptotic features. The two degradation processes are also redundantly required for normal development and viability in this organism. Furthermore, the CES-2-like basic region leucine-zipper (bZip) transcription factor ATF-2, an upstream modulator of the core apoptotic cell death pathway, is able to directly regulate the expression of at least two key autophagy-related genes, bec-1/ATG6 and lgg-1/ATG8. Thus, the two cell death mechanisms share a common method of transcriptional regulation. Together, these results imply that under certain physiological and pathological conditions, autophagy and apoptosis are co-regulated to ensure the proper morphogenesis and survival of the developing organism. The identification of apoptosis and autophagy as compensatory cellular pathways in C. elegans might help us to understand how dysregulated PCD in humans can lead to diverse pathologies, including cancer, neurodegeneration and diabetes.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Animals , Apoptosis/genetics , Autophagy/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , In Situ Nick-End Labeling , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
9.
Autophagy ; 4(3): 330-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18219227

ABSTRACT

Aging is a multifactorial process with many mechanisms contributing to the decline. Mutations decreasing insulin/IGF-1 (insulin-like growth factor-1) or TOR (target of rapamycin) kinase-mediated signaling, mitochondrial activity and food intake each extend life span in divergent animal phyla. Understanding how these genetically distinct mechanisms interact to control longevity is a fundamental and fascinating problem in biology. Here we show that mutational inactivation of autophagy genes, which are involved in the degradation of aberrant, damaged cytoplasmic constituents accumulating in all aging cells, accelerates the rate at which the tissues age in the nematode Caenorhabditis elegans. According to our results Drosophila flies deficient in autophagy are also short-lived. We further demonstrate that reduced activity of autophagy genes suppresses life span extension in mutant nematodes with inherent dietary restriction, aberrant insulin/IGF-1 or TOR signaling, and lowered mitochondrial respiration. These findings suggest that the autophagy gene cascade functions downstream of and is inhibited by different longevity pathways in C. elegans, therefore, their effects converge on autophagy genes to slow down aging and lengthen life span. Thus, autophagy may act as a central regulatory mechanism of animal aging.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Aging/physiology , Animals , Autophagy/genetics , Caenorhabditis elegans Proteins/genetics , Drosophila/genetics , Drosophila/physiology , Insulin/physiology , Insulin-Like Growth Factor I/physiology , Longevity/physiology , Mitochondria/physiology , Mutation , Phosphotransferases (Alcohol Group Acceptor)/physiology , Signal Transduction/genetics , Signal Transduction/physiology
10.
Methods Enzymol ; 451: 467-91, 2008.
Article in English | MEDLINE | ID: mdl-19185736

ABSTRACT

Caenorhabditis elegans has been introduced relatively late into the field of autophagy with no previous results by classical methods. Therefore, it has to be studied in parallel with both traditional electron microscopy and modern molecular approaches. In general, correct identification of autophagic elements by electron microscopy is indispensable to establish a firm basis for our understanding of the process. The principles and the method for identification, applied also for C. elegans, are summarized first in this article, to facilitate their utilization both for further studies and the analysis of new cell types and to support researchers new to electron microscopy techniques. Studying autophagy in the worm by electron microscopy has required the development of special handling and sampling techniques in addition to overcoming the general technical difficulties due to the nature of C. elegans samples. These are described in detail, together with some initial qualitative and quantitative results obtained by them. The feasibility of the presented method is supported by data which show that in continuously fed worms the autophagic compartment is in the lower range of the 10(-2)% order of magnitude of the cytoplasmic volume, while immediately after molting or upon starvation in the second larval period, usually more than a 10-fold increase can be measured. In dauer larvae, individual variation of the autophagic compartment is very high. The predauer stage in daf-2 mutants does not seem to show significant constitutive autophagic activity. Some autophagy-related gene mutants show characteristic ultrastuctural features, such as autophagosomes with membrane abnormalities (unc-51/Atg1) or the hypertrophy of multivesicular bodies (let-512/Vps34, bec-1/Atg6).


Subject(s)
Autophagy/physiology , Caenorhabditis elegans/physiology , Caenorhabditis elegans/ultrastructure , Microscopy, Electron/methods , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cytoplasm/chemistry , Cytoplasm/ultrastructure , Microscopy, Electron/instrumentation , Mutation , Phagosomes/metabolism , Phagosomes/ultrastructure , Recombinant Fusion Proteins , Starvation
11.
Methods Enzymol ; 451: 521-40, 2008.
Article in English | MEDLINE | ID: mdl-19185738

ABSTRACT

Autophagy (cellular self-eating) is a highly regulated, lysosome-mediated catabolic process of eukaryotic cells to segregate by a special membrane and subsequently degrade their own constituents during development or starvation. Electron microscopy analysis reveals autophagic elements in various cell types of the nematode Caenorhabditis elegans, whose genome contains counterparts of several yeast genes involved in autophagy. Genetic manipulation inactivating autophagy-related genes in C. elegans causes defects in development, affects dauer larval morphogenesis, accelerates aging thereby shortening life span, reduces cell size, decreases survival during starvation, promotes apoptotic cell death, and protects neurons from undergoing hyperactive ion channel- or neurotoxin-induced degeneration. These results implicate autophagy in various developmental and cellular functions such as reproductive growth, aging, and cell growth, as well as cell survival and loss. This chapter discusses methods of inactivating C. elegans autophagy genes by RNA interference, testing the resistance of autophagy-deficient nematodes to starvation-induced stress, handling mutants carrying a deletion in the autophagy pathway, and monitoring autophagic activity by using LysoTracker Red dye or reporters labeled with green fluorescent protein. Such methods may be adaptable to identify additional roles of autophagy in development and cellular function, and may also help to detect the intracellular accumulation of autophagy proteins and monitor autophagosome formation.


Subject(s)
Autophagy/genetics , Caenorhabditis elegans/physiology , Animals , Autophagy/physiology , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Longevity/genetics , Mutation , Phagosomes/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Starvation/genetics , Survival Rate
12.
Genetics ; 177(1): 655-60, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17890369

ABSTRACT

Here we show that in the nematode Caenorhabditis elegans mutational inactivation of two autophagy genes unc-51/atg1 and bec-1/atg6/beclin1 results in small body size without affecting cell number. Furthermore, loss-of-function mutations in unc-51 and bec-1 suppress the giant phenotype of mutant animals with aberrant insulin-like growth factor-1 (insulin/IGF-1) or transforming growth factor-beta (TGF-beta) signaling. This function for unc-51 and bec-1 in cell size control and their interaction with these two growth modulatory pathways may represent a link between the hormonal and nutritional regulation of cell growth.


Subject(s)
Autophagy , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/growth & development , Cell Size , Protein Serine-Threonine Kinases/physiology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Mutation , Phenotype , Somatomedins/metabolism , Transforming Growth Factor beta/metabolism , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...