Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Free Radic Biol Med ; 170: 59-69, 2021 07.
Article in English | MEDLINE | ID: mdl-33271282

ABSTRACT

Epigenetic modifications influence gene expression programs ultimately dictating physiological outcomes. In the past decades, an increasing body of work has demonstrated that the enzymes that deposit and/or remove epigenetic marks on DNA or histones use metabolites as substrates or co-factors, rendering the epigenome sensitive to metabolic changes. In this context, acetyl-CoA and α-ketoglutarate have been recognized as critical for epigenetics, impinging on histone marks and nuclear DNA methylation patterns. Given that these metabolites are primarily generated in the mitochondria through the tricarboxylic acid cycle (TCA), the requirement of proper mitochondrial function for maintenance of the epigenetic landscape seems obvious. Nevertheless, it was not until recently when the epigenomic outcomes of mitochondrial dysfunction were tested, revealing mitochondria's far-reaching impact on epigenetics. This review will focus on data that directly tested the role of mitochondria on the epigenetic landscape, the mechanisms by which mitochondrial dysfunction may dysregulate the epigenome and gene expression, and their potential implications to health and disease.


Subject(s)
Epigenesis, Genetic , Epigenome , DNA Methylation , Histones/genetics , Histones/metabolism , Mitochondria/genetics , Mitochondria/metabolism
2.
Int J Mol Sci ; 20(6)2019 Mar 26.
Article in English | MEDLINE | ID: mdl-30917518

ABSTRACT

Telomerase has cellular functions beyond telomere stabilization, including a role in mitochondria. The function of the catalytic component-TERT-in mitochondria is still unknown, but it seems to play a role in the response to oxidative stress. Here, we interrogated the role of the subcellular localization of TERT to the response to hydrogen peroxide (H2O2) treatment. Using normal human fibroblasts (NHF) expressing non-tagged wild type (WT) human TERT (hTERT) or nuclear localization and function (nuchTERT), a mutant that we previously described as being competent in telomere elongation, while not being able to localize to mitochondria, we found the differential activation of autophagy as a function of hTERT's subcellular localization. Specifically, we found that only cells expressing the mutant had significant increases in autophagy markers as a response to H2O2 challenge. Either the reintroduction of the mitochondrial pool of hTERT or the expression of mitochondrially-targeted catalase in mutant cells blunted the autophagic response under oxidative stress. Interestingly, autophagy activation was also associated with decreased levels of mitochondrial DNA damage. Taken together, these results suggest that the loss of hTERT in mitochondria initiates a signaling cascade that allows for cells to adapt to and cope with the lack of mitochondrial telomerase. Such effects also influence the cellular response to oxidative damage.


Subject(s)
Autophagy , Mitochondria/metabolism , Oxidative Stress , Telomerase/metabolism , Cell Line , Fibroblasts/metabolism , Humans , Mutation , Reactive Oxygen Species/metabolism , Telomerase/genetics
3.
Antioxid Redox Signal ; 24(18): 1072-83, 2016 06 20.
Article in English | MEDLINE | ID: mdl-26935406

ABSTRACT

AIMS: Fuchs endothelial corneal dystrophy (FECD), a leading cause of age-related corneal edema requiring transplantation, is characterized by rosette formation of corneal endothelium with ensuing apoptosis. We sought to determine whether excess of mitochondrial reactive oxygen species leads to chronic accumulation of oxidative DNA damage and mitochondrial dysfunction, instigating cell death. RESULTS: We modeled the pathognomonic rosette formation of postmitotic corneal cells by increasing endogenous cellular oxidative stress with menadione (MN) and performed a temporal analysis of its effect in normal (HCEnC, HCECi) and FECD (FECDi) cells and ex vivo specimens. FECDi and FECD ex vivo specimens exhibited extensive mtDNA and nDNA damage as detected by quantitative PCR. Exposure to MN triggered an increase in mitochondrial superoxide levels and led to mtDNA and nDNA damage, while DNA amplification was restored with NAC pretreatment. Furthermore, MN exposure led to a decrease in ΔΨm and adenosine triphosphate levels in normal cells, while FECDi exhibited mitochondrial dysfunction at baseline. Mitochondrial fragmentation and cytochrome c release were detected in FECD tissue and after MN treatment of HCEnCs. Furthermore, cleavage of caspase-9 and caspase-3 followed MN-induced cytochrome c release in HCEnCs. INNOVATION: This study provides the first line of evidence that accumulation of oxidative DNA damage leads to rosette formation, loss of functionally intact mitochondria via fragmentation, and subsequent cell death during postmitotic cell degeneration of ocular tissue. CONCLUSION: MN induced rosette formation, along with mtDNA and nDNA damage, mitochondrial dysfunction, and fragmentation, leading to activation of the intrinsic apoptosis via caspase cleavage and cytochrome c release. Antioxid. Redox Signal. 24, 1072-1083.


Subject(s)
DNA Damage , Fuchs' Endothelial Dystrophy/pathology , Mitochondria/drug effects , Vitamin K 3/toxicity , Apoptosis , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Cytochromes c/metabolism , DNA, Mitochondrial/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/pathology , Oxidative Stress , Rosette Formation
4.
Biomed Res Int ; 2014: 676758, 2014.
Article in English | MEDLINE | ID: mdl-25197655

ABSTRACT

The increased production of reactive oxygen species (ROS) plays a key role in pathogenesis of diabetic complications. ROS are generated by exogenous and endogenous factors such as during hyperglycemia. When ROS production exceeds the detoxification and scavenging capacity of the cell, oxidative stress ensues. Oxidative stress induces DNA damage and when DNA damage exceeds the cellular capacity to repair it, the accumulation of errors can overwhelm the cell resulting in cell death or fixation of genome mutations that can be transmitted to future cell generations. These mutations can lead to and/or play a role in cancer development. This review aims at (i) understanding the types and consequences of DNA damage during hyperglycemic pregnancy; (ii) identifying the biological role of DNA repair during pregnancy, and (iii) proposing clinical interventions to maintain genome integrity. While hyperglycemia can damage the maternal genetic material, the impact of hyperglycemia on fetal cells is still unclear. DNA repair mechanisms may be important to prevent the deleterious effects of hyperglycemia both in mother and in fetus DNA and, as such, prevent the development of diseases in adulthood. Hence, in clinical practice, maternal glycemic control may represent an important point of intervention to prevent the deleterious effects of maternal hyperglycemia to DNA.


Subject(s)
DNA Damage , Fetus/pathology , Hyperglycemia/pathology , DNA Repair , Female , Genomic Instability , Humans , Oxidative Stress , Pregnancy
5.
Nucleic Acids Res ; 40(2): 712-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21937513

ABSTRACT

Human telomerase reverse transcriptase (hTERT) is localized to mitochondria, as well as the nucleus, but details about its biology and function in the organelle remain largely unknown. Here we show, using multiple approaches, that mammalian TERT is mitochondrial, co-purifying with mitochondrial nucleoids and tRNAs. We demonstrate the canonical nuclear RNA [human telomerase RNA (hTR)] is not present in human mitochondria and not required for the mitochondrial effects of telomerase, which nevertheless rely on reverse transcriptase (RT) activity. Using RNA immunoprecipitations from whole cell and in organello, we show that hTERT binds various mitochondrial RNAs, suggesting that RT activity in the organelle is reconstituted with mitochondrial RNAs. In support of this conclusion, TERT drives first strand cDNA synthesis in vitro in the absence of hTR. Finally, we demonstrate that absence of hTERT specifically in mitochondria with maintenance of its nuclear function negatively impacts the organelle. Our data indicate that mitochondrial hTERT works as a hTR-independent reverse transcriptase, and highlight that nuclear and mitochondrial telomerases have different cellular functions. The implications of these findings to both the mitochondrial and telomerase fields are discussed.


Subject(s)
Mitochondria/enzymology , Reverse Transcription , Telomerase/metabolism , Cells, Cultured , DNA, Mitochondrial/isolation & purification , Humans , Membrane Potential, Mitochondrial , Mitochondria/ultrastructure , Protein Transport , RNA/isolation & purification , RNA/metabolism , RNA, Mitochondrial , RNA, Transfer/isolation & purification , Telomerase/isolation & purification
6.
J Nucleic Acids ; 20102010 Sep 21.
Article in English | MEDLINE | ID: mdl-20936168

ABSTRACT

Telomerase is a reverse transcriptase specialized in telomere synthesis. The enzyme is primarily nuclear where it elongates telomeres but recent reports have shown that it also localizes to mitochondria. The function of TERT in mitochondria is largely unknown but the available findings point to a role in mitochondrial DNA metabolism. This paper discusses the available data on mitochondrial telomerase with particular emphasis on its effects upon the organellar DNA.

7.
Hum Mol Genet ; 15(11): 1757-68, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16613901

ABSTRACT

We have previously shown that the protein subunit of telomerase, hTERT, has a bonafide N-terminal mitochondrial targeting sequence, and that ectopic hTERT expression in human cells correlated with increase in mtDNA damage after hydrogen peroxide treatment. In this study, we show, using a loxP hTERT construct, that this increase in mtDNA damage following hydrogen peroxide exposure is dependent on the presence of hTERT itself. Further experiments using a dominant negative hTERT mutant shows that telomerase must be catalytically active to mediate the increase in mtDNA damage. Etoposide, but not methylmethanesulfate, also promotes mtDNA lesions in cells expressing active hTERT, indicating genotoxic specificity in this response. Fibroblasts expressing hTERT not only show a approximately 2-fold increase in mtDNA damage after oxidative stress but also suffer a 10-30-fold increase in apoptotic cell death as assayed by Annexin-V staining, caspase-3 activation and PARP cleavage. Mutations to the N-terminal mitochondrial leader sequence causes a complete loss of mitochondrial targeting without affecting catalytic activity. Cells carrying this mutated hTERT not only have significantly reduced levels of mtDNA damage following hydrogen peroxide treatment, but strikingly also do not shown any loss of viability or cell growth. Thus, localization of hTERT to the mitochondria renders cells more susceptible to oxidative stress-induced mtDNA damage and subsequent cell death, whereas nuclear-targeted hTERT, in the absence of mitochondrial localization, is associated with diminished mtDNA damage, increased cell survival and protection against cellular senescence.


Subject(s)
Apoptosis , DNA Damage , DNA, Mitochondrial/metabolism , Hydrogen Peroxide/pharmacology , Mitochondria/enzymology , Telomerase/biosynthesis , Amino Acid Sequence , Caspase 3 , Caspases/metabolism , Enzyme Activation , Etoposide/pharmacology , Fibroblasts/metabolism , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Molecular Sequence Data , Poly(ADP-ribose) Polymerases/metabolism , Sequence Homology, Amino Acid
8.
Aging Cell ; 3(6): 399-411, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15569357

ABSTRACT

Telomerase is often re-activated in human cancers and is widely used to immortalize cells in culture. In addition to the maintenance of telomeres, telomerase has been implicated in cell proliferation, genomic instability and apoptosis. Here we show that human telomerase reverse transcriptase (hTERT) is targeted to the mitochondria by an N-terminal leader sequence, and that mitochondrial extracts contain telomerase activity. In seven different human cell lines, mitochondrial telomerase increases hydrogen-peroxide-mediated mitochondrial DNA damage. hTERT expression did not alter the rate of hydrogen peroxide breakdown or endogenous cellular levels. Because the damaging effects of hydrogen peroxide are mediated by divalent metal ions (Fenton chemistry), we examined the levels of bioavailable metals. In all cases, higher levels of chelatable metals were found in hTERT-expressing cells. These results suggest that mitochondrial telomerase sensitizes cells to oxidative stress, which can lead to apoptotic cell death, and imply a novel function of telomerase in mitochondrial DNA transactions.


Subject(s)
DNA Damage/drug effects , DNA, Mitochondrial/drug effects , Mitochondria/metabolism , Telomerase/metabolism , Algorithms , Amino Acid Sequence , Cell Death/drug effects , Cell Line , DNA, Mitochondrial/metabolism , DNA-Binding Proteins , Free Radicals/metabolism , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Molecular Sequence Data , Sensitivity and Specificity , Telomerase/genetics
9.
J Biol Chem ; 278(3): 1728-34, 2003 Jan 17.
Article in English | MEDLINE | ID: mdl-12424245

ABSTRACT

In order to understand the molecular events following oxidative stress, which lead to persistence of lesions in the mtDNA, experiments were performed on normal human fibroblast (NHF) expressing human telomerase reverse transcriptase (hTERT). The formation and repair of H(2)O(2)-induced DNA lesions were examined using quantitative PCR. It was found that NHF hTERTs show extensive mtDNA damage ( approximately 4 lesions/10 kb) after exposure to 200 microm H(2)O(2), which is partially repaired during a recovery period of 6 h. At the same time, the nDNA seemed to be completely resistant to damage. Cell sorting experiments revealed persistent mtDNA damage at 24 h only in the fraction of cells with low mitochondrial membrane potential (Delta Psi m). Further analysis also showed increased production of H(2)O(2) by these cells, which subsequently undergo apoptosis. This work supports a hypothesis for a feed-forward cascade of reactive oxygen species generation and mtDNA damage and also suggested a possible mechanism for persistence of lesions in the mtDNA involving a drop in Delta Psi m, compromised protein import, secondary reactive oxygen species generation, and loss of repair capacity.


Subject(s)
Apoptosis , DNA Damage , DNA, Mitochondrial/drug effects , Intracellular Membranes/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Cell Separation , Cells, Cultured , Cysteine Endopeptidases/metabolism , DNA, Mitochondrial/genetics , Enzyme Activation , Flow Cytometry , Humans , Hydrogen Peroxide/pharmacology , Intracellular Membranes/physiology , Mitochondria/enzymology , Mitochondria/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...