Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(12)2023 Jun 18.
Article in English | MEDLINE | ID: mdl-37373453

ABSTRACT

Chemically modified nucleic acid bases are sources of genomic instability and mutations but may also regulate gene expression as epigenetic or epitranscriptomic modifications. Depending on the cellular context, they can have vastly diverse impacts on cells, from mutagenesis or cytotoxicity to changing cell fate by regulating chromatin organisation and gene expression. Identical chemical modifications exerting different functions pose a challenge for the cell's DNA repair machinery, as it needs to accurately distinguish between epigenetic marks and DNA damage to ensure proper repair and maintenance of (epi)genomic integrity. The specificity and selectivity of the recognition of these modified bases relies on DNA glycosylases, which acts as DNA damage, or more correctly, as modified bases sensors for the base excision repair (BER) pathway. Here, we will illustrate this duality by summarizing the role of uracil-DNA glycosylases, with particular attention to SMUG1, in the regulation of the epigenetic landscape as active regulators of gene expression and chromatin remodelling. We will also describe how epigenetic marks, with a special focus on 5-hydroxymethyluracil, can affect the damage susceptibility of nucleic acids and conversely how DNA damage can induce changes in the epigenetic landscape by altering the pattern of DNA methylation and chromatin structure.


Subject(s)
DNA Damage , DNA Repair , Mutation , DNA Methylation
2.
Brain ; 146(8): 3513-3527, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36917474

ABSTRACT

RNA polymerase I transcribes ribosomal DNA to produce precursor 47S rRNA. Post-transcriptional processing of this rRNA generates mature 28S, 18S and 5.8S rRNAs, which form the ribosomes, together with 5S rRNA, assembly factors and ribosomal proteins. We previously reported a homozygous variant in the catalytic subunit of RNA polymerase I, POLR1A, in two brothers with leukodystrophy and progressive course. However, the disease mechanism remained unknown. In this report, we describe another missense variant POLR1A NM_015425.3:c.1925C>A; p.(Thr642Asn) in homozygosity in two unrelated patients. Patient 1 was a 16-year-old male and Patient 2 was a 2-year-old female. Both patients manifested neurological deficits, with brain MRIs showing hypomyelinating leukodystrophy and cerebellar atrophy; and in Patient 1 additionally with hypointensity of globi pallidi and small volume of the basal ganglia. Patient 1 had progressive disease course, leading to death at the age of 16.5 years. Extensive in vitro experiments in fibroblasts from Patient 1 documented that the mutated POLR1A led to aberrant rRNA processing and degradation, and abnormal nucleolar homeostasis. Proteomics data analyses and further in vitro experiments documented abnormal protein homeostasis, and endoplasmic reticulum stress responses. We confirm that POLR1A biallelic variants cause neurodegenerative disease, expand the knowledge of the clinical phenotype of the disorder, and provide evidence for possible pathological mechanisms leading to POLR1A-related leukodystrophy.


Subject(s)
Neurodegenerative Diseases , RNA Polymerase I , Male , Female , Humans , RNA Polymerase I/genetics , RNA Polymerase I/metabolism , Neurodegenerative Diseases/genetics , Proteostasis , RNA, Ribosomal/metabolism , Ribosomes , RNA Processing, Post-Transcriptional
3.
Aging (Albany NY) ; 14(21): 8661-8687, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36367773

ABSTRACT

There is accumulating evidence that interfering with the basic aging mechanisms can enhance healthy longevity. The interventional/therapeutic strategies targeting multiple aging hallmarks could be more effective than targeting one hallmark. While health-promoting qualities of marine oils have been extensively studied, the underlying molecular mechanisms are not fully understood. Lipid extracts from Antarctic krill are rich in long-chain omega-3 fatty acids choline, and astaxanthin. Here, we used C. elegans and human cells to investigate whether krill oil promotes healthy aging. In a C. elegans model of Parkinson´s disease, we show that krill oil protects dopaminergic neurons from aging-related degeneration, decreases alpha-synuclein aggregation, and improves dopamine-dependent behavior and cognition. Krill oil rewires distinct gene expression programs that contribute to attenuating several aging hallmarks, including oxidative stress, proteotoxic stress, senescence, genomic instability, and mitochondrial dysfunction. Mechanistically, krill oil increases neuronal resilience through temporal transcriptome rewiring to promote anti-oxidative stress and anti-inflammation via healthspan regulating transcription factors such as SNK-1. Moreover, krill oil promotes dopaminergic neuron survival through regulation of synaptic transmission and neuronal functions via PBO-2 and RIM-1. Collectively, krill oil rewires global gene expression programs and promotes healthy aging via abrogating multiple aging hallmarks, suggesting directions for further pre-clinical and clinical explorations.


Subject(s)
Dopaminergic Neurons , Euphausiacea , Humans , Animals , Transcriptome , Caenorhabditis elegans , Plant Oils , Dopamine
4.
DNA Repair (Amst) ; 120: 103410, 2022 12.
Article in English | MEDLINE | ID: mdl-36244177

ABSTRACT

Fatty liver diseases are a major health threat across the western world, leading to cirrhosis and premature morbidity and mortality. Recently, a correlation between the base excision repair enzyme SMUG1 and metabolic homeostasis was identified. As the molecular mechanisms remain unknown, we exploited a SMUG1-knockout mouse model to gain insights into this association by characterizing the liver phenotype in young vs old SMUG1-null mice. We observed increased weight and fat content in one-year old animals, with altered activity of enzymes important for fatty acids influx and uptake. Consistently, lipidomic profiling showed accumulation of free fatty acids and triglycerides in SMUG1-null livers. Old SMUG1-knockout mice also displayed increased hepatocyte senescence and DNA damage at telomeres. Interestingly, RNA sequencing revealed widespread changes in the expression of lipid metabolic genes already in three months old animals. In summary, SMUG1 modulates fat metabolism favouring net lipogenesis and resulting in development of a fatty liver phenotype.


Subject(s)
Fatty Liver , Uracil-DNA Glycosidase , Mice , Animals , Uracil-DNA Glycosidase/metabolism , Fatty Liver/metabolism , Mice, Knockout , Phenotype , Homeostasis , Liver/metabolism
5.
Nucleic Acids Res ; 50(18): 10449-10468, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36156150

ABSTRACT

Single-strand selective uracil-DNA glycosylase 1 (SMUG1) initiates base excision repair (BER) of uracil and oxidized pyrimidines. SMUG1 status has been associated with cancer risk and therapeutic response in breast carcinomas and other cancer types. However, SMUG1 is a multifunctional protein involved, not only, in BER but also in RNA quality control, and its function in cancer cells is unclear. Here we identify several novel SMUG1 interaction partners that functions in many biological processes relevant for cancer development and treatment response. Based on this, we hypothesized that the dominating function of SMUG1 in cancer might be ascribed to functions other than BER. We define a bad prognosis signature for SMUG1 by mapping out the SMUG1 interaction network and found that high expression of genes in the bad prognosis network correlated with lower survival probability in ER+ breast cancer. Interestingly, we identified hsa-let-7b-5p microRNA as an upstream regulator of the SMUG1 interactome. Expression of SMUG1 and hsa-let-7b-5p were negatively correlated in breast cancer and we found an inhibitory auto-regulatory loop between SMUG1 and hsa-let-7b-5p in the MCF7 breast cancer cells. We conclude that SMUG1 functions in a gene regulatory network that influence the survival and treatment response in several cancers.


Subject(s)
Breast Neoplasms , MicroRNAs , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , MicroRNAs/genetics , Prognosis , Uracil/metabolism , Uracil-DNA Glycosidase/genetics
6.
DNA Repair (Amst) ; 99: 103051, 2021 03.
Article in English | MEDLINE | ID: mdl-33540225

ABSTRACT

The integrity of the genetic information is continuously challenged by numerous genotoxic insults, most frequently in the form of oxidation, alkylation or deamination of the bases that result in DNA damage. These damages compromise the fidelity of the replication, and interfere with the progression and function of the transcription machineries. The DNA damage response (DDR) comprises a series of strategies to deal with DNA damage, including transient transcriptional inhibition, activation of DNA repair pathways and chromatin remodeling. Coordinated control of transcription and DNA repair is required to safeguardi cellular functions and identities. Here, we address the cellular responses to endogenous DNA damage, with a particular focus on the role of DNA glycosylases and the Base Excision Repair (BER) pathway, in conjunction with the DDR factors, in responding to DNA damage during the transcription process. We will also discuss functions of newly identified epigenetic and regulatory marks, such as 5-hydroxymethylcytosine and its oxidative products and 8-oxoguanine, that were previously considered only as DNA damages. In light of these resultsthe classical perception of DNA damage as detrimental for cellular processes are changing. and a picture emerges whereDNA glycosylases act as dynamic regulators of transcription, placing them at the intersection of DNA repair and gene expression modulation.


Subject(s)
DNA Damage , DNA Glycosylases/metabolism , DNA Repair , Epigenesis, Genetic , 5-Methylcytosine/analogs & derivatives , Animals , DNA/metabolism , Gene Expression Regulation , Guanine/analogs & derivatives , Humans
7.
Biomolecules ; 11(1)2021 01 08.
Article in English | MEDLINE | ID: mdl-33430019

ABSTRACT

RNA modifications are essential for proper RNA processing, quality control, and maturation steps. In the last decade, some eukaryotic DNA repair enzymes have been shown to have an ability to recognize and process modified RNA substrates and thereby contribute to RNA surveillance. Single-strand-selective monofunctional uracil-DNA glycosylase 1 (SMUG1) is a base excision repair enzyme that not only recognizes and removes uracil and oxidized pyrimidines from DNA but is also able to process modified RNA substrates. SMUG1 interacts with the pseudouridine synthase dyskerin (DKC1), an enzyme essential for the correct assembly of small nucleolar ribonucleoproteins (snRNPs) and ribosomal RNA (rRNA) processing. Here, we review rRNA modifications and RNA quality control mechanisms in general and discuss the specific function of SMUG1 in rRNA metabolism. Cells lacking SMUG1 have elevated levels of immature rRNA molecules and accumulation of 5-hydroxymethyluridine (5hmU) in mature rRNA. SMUG1 may be required for post-transcriptional regulation and quality control of rRNAs, partly by regulating rRNA and stability.


Subject(s)
RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , Uracil-DNA Glycosidase/metabolism , Animals , Humans , Models, Molecular , RNA Stability , RNA, Ribosomal/chemistry
8.
Biomolecules ; 10(4)2020 03 31.
Article in English | MEDLINE | ID: mdl-32244430

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1), the main mammalian AP-endonuclease for the resolution of DNA damages through the base excision repair (BER) pathway, acts as a multifunctional protein in different key cellular processes. The signals to ensure temporo-spatial regulation of APE1 towards a specific function are still a matter of debate. Several studies have suggested that post-translational modifications (PTMs) act as dynamic molecular mechanisms for controlling APE1 functionality. Interestingly, the N-terminal region of APE1 is a disordered portion functioning as an interface for protein binding, as an acceptor site for PTMs and as a target of proteolytic cleavage. We previously demonstrated a cytoplasmic accumulation of truncated APE1 in acute myeloid leukemia (AML) cells in association with a mutated form of nucleophosmin having aberrant cytoplasmic localization (NPM1c+). Here, we mapped the proteolytic sites of APE1 in AML cells at Lys31 and Lys32 and showed that substitution of Lys27, 31, 32 and 35 with alanine impairs proteolysis. We found that the loss of the APE1 N-terminal domain in AML cells is dependent on the proteasome, but not on granzyme A/K as described previously. The present work identified the proteasome as a contributing machinery involved in APE1 cleavage in AML cells, suggesting that acetylation can modulate this process.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Leukemia, Myeloid, Acute/pathology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Acetylation , Amino Acid Sequence , Cell Line, Tumor , Humans , Protein Domains
9.
Mol Cell Oncol ; 6(6): e1670489, 2019.
Article in English | MEDLINE | ID: mdl-31692866

ABSTRACT

Disturbances in telomere maintenance are common in cancer. We recently showed that Single-strand-selective monofunctional uracil-DNA glycosylase 1 (SMUG1) promotes telomere homeostasis by regulating the stability of the telomeric RNA component (hTERC). SMUG1-mediated recognition of base modifications may function in a regulated process serving to fine-tune the levels of hTERC.

10.
Cell Rep ; 28(7): 1690-1702.e10, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412240

ABSTRACT

Telomerase biogenesis is a complex process where several steps remain poorly understood. Single-strand-selective uracil-DNA glycosylase (SMUG1) associates with the DKC1-containing H/ACA ribonucleoprotein complex, which is essential for telomerase biogenesis. Herein, we show that SMUG1 interacts with the telomeric RNA component (hTERC) and is required for co-transcriptional processing of the nascent transcript into mature hTERC. We demonstrate that SMUG1 regulates the presence of base modifications in hTERC, in a region between the CR4/CR5 domain and the H box. Increased levels of hTERC base modifications are accompanied by reduced DKC1 binding. Loss of SMUG1 leads to an imbalance between mature hTERC and its processing intermediates, leading to the accumulation of 3'-polyadenylated and 3'-extended intermediates that are degraded in an EXOSC10-independent RNA degradation pathway. Consequently, SMUG1-deprived cells exhibit telomerase deficiency, leading to impaired bone marrow proliferation in Smug1-knockout mice.


Subject(s)
RNA Processing, Post-Transcriptional , RNA/physiology , Telomerase/metabolism , Telomere/physiology , Uracil-DNA Glycosidase/metabolism , Animals , Exoribonucleases/genetics , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Female , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Telomerase/genetics , Telomerase/physiology , Uracil-DNA Glycosidase/genetics , Uracil-DNA Glycosidase/physiology
11.
Nat Commun ; 8(1): 797, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28986522

ABSTRACT

Mammalian apurinic/apyrimidinic endonuclease 1 is a DNA repair enzyme involved in genome stability and expression of genes involved in oxidative stress responses, tumor progression and chemoresistance. However, the molecular mechanisms underlying the role of apurinic/apyrimidinic endonuclease 1 in these processes are still unclear. Recent findings point to a novel role of apurinic/apyrimidinic endonuclease 1 in RNA metabolism. Through the characterization of the interactomes of apurinic/apyrimidinic endonuclease 1 with RNA and other proteins, we demonstrate here a role for apurinic/apyrimidinic endonuclease 1 in pri-miRNA processing and stability via association with the DROSHA-processing complex during genotoxic stress. We also show that endonuclease activity of apurinic/apyrimidinic endonuclease 1 is required for the processing of miR-221/222 in regulating expression of the tumor suppressor PTEN. Analysis of a cohort of different cancers supports the relevance of our findings for tumor biology. We also show that apurinic/apyrimidinic endonuclease 1 participates in RNA-interactomes and protein-interactomes involved in cancer development, thus indicating an unsuspected post-transcriptional effect on cancer genes.APE1 plays an important role in the cellular response to oxidative stress, and mutations are linked to tumor progression and chemoresistance. Here, the authors characterize the interactions of APE1 with RNA and demonstrate a role in microRNA processing.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , MicroRNAs/metabolism , Neoplasms/metabolism , Brain Neoplasms/metabolism , Breast Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , HCT116 Cells , HeLa Cells , Humans , MCF-7 Cells , Male , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Uterine Cervical Neoplasms/metabolism
12.
Sci Rep ; 7(1): 7199, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28775312

ABSTRACT

Both a DNA lesion and an intermediate for antibody maturation, uracil is primarily processed by base excision repair (BER), either initiated by uracil-DNA glycosylase (UNG) or by single-strand selective monofunctional uracil DNA glycosylase (SMUG1). The relative in vivo contributions of each glycosylase remain elusive. To assess the impact of SMUG1 deficiency, we measured uracil and 5-hydroxymethyluracil, another SMUG1 substrate, in Smug1 -/- mice. We found that 5-hydroxymethyluracil accumulated in Smug1 -/- tissues and correlated with 5-hydroxymethylcytosine levels. The highest increase was found in brain, which contained about 26-fold higher genomic 5-hydroxymethyluracil levels than the wild type. Smug1 -/- mice did not accumulate uracil in their genome and Ung -/- mice showed slightly elevated uracil levels. Contrastingly, Ung -/- Smug1 -/- mice showed a synergistic increase in uracil levels with up to 25-fold higher uracil levels than wild type. Whole genome sequencing of UNG/SMUG1-deficient tumours revealed that combined UNG and SMUG1 deficiency leads to the accumulation of mutations, primarily C to T transitions within CpG sequences. This unexpected sequence bias suggests that CpG dinucleotides are intrinsically more mutation prone. In conclusion, we showed that SMUG1 efficiently prevent genomic uracil accumulation, even in the presence of UNG, and identified mutational signatures associated with combined UNG and SMUG1 deficiency.


Subject(s)
Cytosine/metabolism , Dinucleoside Phosphates/metabolism , Uracil-DNA Glycosidase/deficiency , Uracil/metabolism , Animals , CpG Islands , Deamination , Genome , Genomics/methods , Mice , Mice, Knockout , Mutation
13.
Oncotarget ; 7(37): 59129-59143, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27494862

ABSTRACT

Nucleophosmin (NPM1) is a multifunctional protein that is implicated in the pathogenesis of several human malignancies. To gain insight into the role of isolated fragments of NPM1 in its biological activities, we dissected the C-terminal domain (CTD) into its helical fragments. Here we focus the attention on the third helix of the NPM1-CTD in its wild-type (H3 wt) and AML-mutated (H3 mutA and H3 mutE) sequences. Conformational studies, by means of CD and NMR spectroscopies, showed that the H3 wt peptide was partially endowed with an α-helical structure, but the AML-sequences exhibited a lower content of this conformation, particularly the H3 mutA peptide. Thioflavin T assays showed that the H3 mutE and the H3 mutA peptides displayed a significant aggregation propensity that was confirmed by CD and DLS assays. In addition, we found that the H3 mutE and H3 mutA peptides, unlike the H3 wt, were moderately and highly toxic, respectively, when exposed to human neuroblastoma cells. Cellular localization experiments confirmed that the mutated sequences hamper their nucleolar accumulation, and more importantly, that the helical conformation of the H3 region is crucial for such a localization.


Subject(s)
Cell Nucleus/metabolism , Cytosol/metabolism , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Apoptosis , Cell Line, Tumor , Humans , Nucleophosmin , Peptide Fragments , Protein Aggregation, Pathological , Protein Conformation , Protein Stability , Protein Transport
14.
Oncotarget ; 7(18): 26293-306, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27050370

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1) is the main mammalian AP-endonuclease responsible for the repair of endogenous DNA damage through the base excision repair (BER) pathway. Molecular epidemiological studies have identified several genetic variants associated with human diseases, but a well-defined functional connection between mutations in APE1 and disease development is lacking. In order to understand the biological consequences of APE1 genetic mutations, we examined the molecular and cellular consequences of the selective expression of four non-synonymous APE1 variants (L104R, R237C, D148E and D283G) in human cells. We found that D283G, L104R and R237C variants have reduced endonuclease activity and impaired ability to associate with XRCC1 and DNA polymerase ß, which are enzymes acting downstream of APE1 in the BER pathway. Complementation experiments performed in cells, where endogenous APE1 had been silenced by shRNA, showed that the expression of these variants resulted in increased phosphorylation of histone H2Ax and augmented levels of poly(ADP-ribosyl)ated (PAR) proteins. Persistent activation of DNA damage response markers was accompanied by growth defects likely due to combined apoptotic and autophagic processes. These phenotypes were observed in the absence of exogenous stressors, suggesting that chronic replication stress elicited by the BER defect may lead to a chronic activation of the DNA damage response. Hence, our data reinforce the concept that non-synonymous APE1 variants present in the human population may act as cancer susceptibility alleles.


Subject(s)
DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Genetic Predisposition to Disease/genetics , Neoplasms/genetics , Cell Proliferation/genetics , Genotype , HeLa Cells , Humans , Polymorphism, Single Nucleotide
15.
Mol Biol Cell ; 25(10): 1641-52, 2014 May.
Article in English | MEDLINE | ID: mdl-24648491

ABSTRACT

Nucleophosmin (NPM1) is a multifunctional protein that controls cell growth and genome stability via a mechanism that involves nucleolar-cytoplasmic shuttling. It is clear that NPM1 also contributes to the DNA damage response, yet its exact function is poorly understood. We recently linked NPM1 expression to the functional activation of the major abasic endonuclease in mammalian base excision repair (BER), apurinic/apyrimidinic endonuclease 1 (APE1). Here we unveil a novel role for NPM1 as a modulator of the whole BER pathway by 1) controlling BER protein levels, 2) regulating total BER capacity, and 3) modulating the nucleolar localization of several BER enzymes. We find that cell treatment with the genotoxin cisplatin leads to concurrent relocalization of NPM1 and BER components from nucleoli to the nucleoplasm, and cellular experiments targeting APE1 suggest a role for the redistribution of nucleolar BER factors in determining cisplatin toxicity. Finally, based on the use of APE1 as a representative protein of the BER pathway, our data suggest a function for BER proteins in the regulation of ribogenesis.


Subject(s)
DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Nuclear Proteins/genetics , Animals , Cell Nucleolus/genetics , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA Damage/genetics , DNA Ligase ATP , DNA Ligases/biosynthesis , DNA Ligases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Doxycycline/pharmacology , Flap Endonucleases/biosynthesis , Flap Endonucleases/metabolism , HeLa Cells , Humans , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/metabolism , Nucleophosmin , Protein Transport/genetics , RNA Interference , RNA, Small Interfering , Ribosomes/genetics , Tumor Suppressor Protein p53/genetics
16.
Antioxid Redox Signal ; 20(4): 621-39, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23879289

ABSTRACT

SIGNIFICANCE: An emerging concept in DNA repair mechanisms is the evidence that some key enzymes, besides their role in the maintenance of genome stability, display also unexpected noncanonical functions associated with RNA metabolism in specific subcellular districts (e.g., nucleoli). During the evolution of these key enzymes, the acquisition of unfolded domains significantly amplified the possibility to interact with different partners and substrates, possibly explaining their phylogenetic gain of functions. RECENT ADVANCES: After nucleolar stress or DNA damage, many DNA repair proteins can freely relocalize from nucleoli to the nucleoplasm. This process may represent a surveillance mechanism to monitor the synthesis and correct assembly of ribosomal units affecting cell cycle progression or inducing p53-mediated apoptosis or senescence. CRITICAL ISSUES: A paradigm for this kind of regulation is represented by some enzymes of the DNA base excision repair (BER) pathway, such as apurinic/apyrimidinic endonuclease 1 (APE1). In this review, the role of the nucleolus and the noncanonical functions of the APE1 protein are discussed in light of their possible implications in human pathologies. FUTURE DIRECTIONS: A productive cross-talk between DNA repair enzymes and proteins involved in RNA metabolism seems reasonable as the nucleolus is emerging as a dynamic functional hub that coordinates cell growth arrest and DNA repair mechanisms. These findings will drive further analyses on other BER proteins and might imply that nucleic acid processing enzymes are more versatile than originally thought having evolved DNA-targeted functions after a previous life in the early RNA world.


Subject(s)
Cell Nucleolus/enzymology , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Animals , Cell Nucleolus/physiology , DNA Damage , DNA Repair Enzymes/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Nuclear Proteins/metabolism , Nucleophosmin , Protein Structure, Tertiary
17.
Mol Biol Cell ; 25(4): 532-47, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24356447

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein contributing to genome stability via repair of DNA lesions via the base excision repair pathway. It also plays a role in gene expression regulation and RNA metabolism. Another, poorly characterized function is its ability to bind to negative calcium responsive elements (nCaRE) of some gene promoters. The presence of many functional nCaRE sequences regulating gene transcription can be envisioned, given their conservation within ALU repeats. To look for functional nCaRE sequences within the human genome, we performed bioinformatic analyses and identified 57 genes potentially regulated by APE1. We focused on sirtuin-1 (SIRT1) deacetylase due to its involvement in cell stress, including senescence, apoptosis, and tumorigenesis, and its role in the deacetylation of APE1 after genotoxic stress. The human SIRT1 promoter presents two nCaRE elements stably bound by APE1 through its N-terminus. We demonstrate that APE1 is part of a multiprotein complex including hOGG1, Ku70, and RNA Pol II, which is recruited on SIRT1 promoter to regulate SIRT1 gene functions during early response to oxidative stress. These findings provide new insights into the role of nCaRE sequences in the transcriptional regulation of mammalian genes.


Subject(s)
DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Gene Expression Regulation/drug effects , Genome, Human , Response Elements , Sirtuin 1/genetics , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Base Sequence , Binding Sites , Computational Biology , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Ku Autoantigen , Methyl Methanesulfonate/pharmacology , Molecular Sequence Annotation , Molecular Sequence Data , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sirtuin 1/metabolism , Transcription, Genetic
18.
Biochem J ; 452(3): 545-57, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23544830

ABSTRACT

The hAPE1 (human apurinic/apyrimidinic endonuclease 1) is an essential enzyme, being the main abasic endonuclease in higher eukaryotes. However, there is strong evidence to show that hAPE1 can directly bind specific gene promoters, thus modulating their transcriptional activity, even in the absence of specific DNA damage. Recent findings, moreover, suggest a role for hAPE1 in RNA processing, which is modulated by the interaction with NPM1 (nucleophosmin). Independent domains account for many activities of hAPE1; however, whereas the endonuclease and the redox-active portions of the protein are well characterized, a better understanding of the role of the unstructured N-terminal region is needed. In the present study, we characterized the requirements for the interaction of hAPE1 with NPM1 and undamaged nucleic acids. We show that DNA/RNA secondary structure has an impact on hAPE1 binding in the absence of damage. Biochemical studies, using the isolated N-terminal region of the protein, reveal that the hAPE1 N-terminal domain represents an evolutionary gain of function, since its composition affects the protein's stability and ability to interact with both nucleic acids and NPM1. Although required, however, this region is not sufficient itself to stably interact with DNA or NPM1.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Nuclear Proteins/chemistry , Nucleic Acids/chemistry , Amino Acid Sequence , Animals , DNA/genetics , DNA/metabolism , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Nucleic Acids/genetics , Nucleophosmin , Protein Binding/genetics , RNA/genetics , RNA/metabolism , Sequence Homology , Zebrafish
19.
Mol Biol Cell ; 23(20): 4079-96, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918947

ABSTRACT

Apurinic/apyrimidinic endonuclease 1 (APE1) is the main abasic endonuclease in the base excision repair (BER) pathway of DNA lesions caused by oxidation/alkylation in mammalian cells; within nucleoli it interacts with nucleophosmin and rRNA through N-terminal Lys residues, some of which (K(27)/K(31)/K(32)/K(35)) may undergo acetylation in vivo. Here we study the functional role of these modifications during genotoxic damage and their in vivo relevance. We demonstrate that cells expressing a specific K-to-A multiple mutant are APE1 nucleolar deficient and are more resistant to genotoxic treatment than those expressing the wild type, although they show impaired proliferation. Of interest, we find that genotoxic treatment induces acetylation at these K residues. We also find that the charged status of K(27)/K(31)/K(32)/K(35) modulates acetylation at K(6)/K(7) residues that are known to be involved in the coordination of BER activity through a mechanism regulated by the sirtuin 1 deacetylase. Of note, structural studies show that acetylation at K(27)/K(31)/K(32)/K(35) may account for local conformational changes on APE1 protein structure. These results highlight the emerging role of acetylation of critical Lys residues in regulating APE1 functions. They also suggest the existence of cross-talk between different Lys residues of APE1 occurring upon genotoxic damage, which may modulate APE1 subnuclear distribution and enzymatic activity in vivo.


Subject(s)
Cell Nucleolus/enzymology , DNA Damage , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Lysine/metabolism , Acetylation , Cell Proliferation , Enzyme Stability , HeLa Cells , Humans , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Protein Binding , Protein Conformation , Protein Transport , RNA, Ribosomal/metabolism , Sirtuin 1/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL