Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Biol Chem ; 295(40): 13902-13913, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32763971

RESUMO

Trinucleotide repeat (TNR) expansion and deletion are responsible for over 40 neurodegenerative diseases and associated with cancer. TNRs can undergo somatic instability that is mediated by DNA damage and repair and gene transcription. Recent studies have pointed toward a role for R-loops in causing TNR expansion and deletion, and it has been shown that base excision repair (BER) can result in CAG repeat deletion from R-loops in yeast. However, it remains unknown how BER in R-loops can mediate TNR instability. In this study, using biochemical approaches, we examined BER enzymatic activities and their influence on TNR R-loops. We found that AP endonuclease 1 incised an abasic site on the nontemplate strand of a TNR R-loop, creating a double-flap intermediate containing an RNA:DNA hybrid that subsequently inhibited polymerase ß (pol ß) synthesis of TNRs. This stimulated flap endonuclease 1 (FEN1) cleavage of TNRs engaged in an R-loop. Moreover, we showed that FEN1 also efficiently cleaved the RNA strand, facilitating pol ß loop/hairpin bypass synthesis and the resolution of TNR R-loops through BER. Consequently, this resulted in fewer TNRs synthesized by pol ß than those removed by FEN1, thereby leading to repeat deletion. Our results indicate that TNR R-loops preferentially lead to repeat deletion during BER by disrupting the balance between the addition and removal of TNRs. Our discoveries open a new avenue for the treatment and prevention of repeat expansion diseases and cancer.


Assuntos
DNA Polimerase beta/química , Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/química , Endonucleases Flap/química , Estruturas R-Loop , Repetições de Trinucleotídeos , Humanos
2.
Analyst ; 145(14): 4852-4859, 2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32542257

RESUMO

Glass nanopipettes have shown promise for applications in single-cell manipulation, analysis, and imaging. In recent years, plasmonic nanopipettes have been developed to enable surface-enhanced Raman spectroscopy (SERS) measurements for single-cell analysis. In this work, we developed a SERS-active nanopipette that can be used to perform long-term and reliable intracellular analysis of single living cells with minimal damage, which is achieved by optimizing the nanopipette geometry and the surface density of the gold nanoparticle (AuNP) layer at the nanopipette tip. To demonstrate its ability in single-cell analysis, we used the nanopipette for intracellular pH sensing. Intracellular pH (pHi) is vital to cells as it influences cell function and behavior and pathological conditions. The pH sensitivity was realized by simply modifying the AuNP layer with the pH reporter molecule 4-mercaptobenzoic acid. With a response time of less than 5 seconds, the pH sensing range is from 6.0 to 8.0 and the maximum sensitivity is 0.2 pH units. We monitored the pHi change of individual HeLa and fibroblast cells, triggered by the extracellular pH (pHe) change. The HeLa cancer cells can better resist pHe change and adapt to the weak acidic environment. Plasmonic nanopipettes can be further developed to monitor other intracellular biomarkers.


Assuntos
Ouro , Nanopartículas Metálicas , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Análise Espectral Raman
3.
Nucleic Acids Res ; 46(17): 8940-8952, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30085293

RESUMO

Trinucleotide repeat (TNR) instability is associated with over 42 neurodegenerative diseases and cancer, for which the molecular mechanisms remain to be elucidated. We have shown that the DNA base excision repair (BER) pathway and its central component, DNA polymerase ß (pol ß), in particular, its polymerase activity plays an active role in regulating somatic TNR instability. Herein, we revealed a unique role of the pol ß dRP lyase in preventing somatic TNR instability. We found that deficiency of pol ß deoxyribose phosphate (dRP) lyase activity locked the pol ß dRP lyase domain to a dRP group, and this 'tethered' pol ß to its template forcing the polymerase to perform a processive DNA synthesis. This subsequently promoted DNA strand slippage allowing pol ß to skip over a template loop and causing TNR deletion. We showed that the effects were eliminated by complementation of the dRP lyase deficiency with wild-type pol ß protein. The results indicate that pol ß dRP lyase activity restrained the pol ß-dRP interaction to suppress a pol ß processive DNA synthesis, thereby preventing TNR deletion. This further implicates a potential of pol ß dRP lyase inhibition as a novel treatment of TNR-expansion diseases.


Assuntos
DNA Polimerase beta/genética , Reparo do DNA , Fósforo-Oxigênio Liases/genética , Expansão das Repetições de Trinucleotídeos , Repetições de Trinucleotídeos , Animais , Sistema Livre de Células/química , Sistema Livre de Células/metabolismo , DNA/biossíntese , DNA/genética , DNA Polimerase beta/metabolismo , Replicação do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Fibroblastos/química , Fibroblastos/metabolismo , Endonucleases Flap/genética , Endonucleases Flap/metabolismo , Regulação da Expressão Gênica , Teste de Complementação Genética , Instabilidade Genômica , Humanos , Camundongos , Oligonucleotídeos/síntese química , Oligonucleotídeos/metabolismo , Fósforo-Oxigênio Liases/metabolismo
4.
Molecules ; 24(21)2019 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-31717862

RESUMO

Cells must faithfully duplicate their DNA in the genome to pass their genetic information to the daughter cells. To maintain genomic stability and integrity, double-strand DNA has to be replicated in a strictly regulated manner, ensuring the accuracy of its copy number, integrity and epigenetic modifications. However, DNA is constantly under the attack of DNA damage, among which oxidative DNA damage is the one that most frequently occurs, and can alter the accuracy of DNA replication, integrity and epigenetic features, resulting in DNA replication stress and subsequent genome and epigenome instability. In this review, we summarize DNA damage-induced replication stress, the formation of DNA secondary structures, peculiar epigenetic modifications and cellular responses to the stress and their impact on the instability of the genome and epigenome mainly in eukaryotic cells.


Assuntos
Dano ao DNA/genética , Epigenoma/genética , DNA/genética , Metilação de DNA/genética , Metilação de DNA/fisiologia , Replicação do DNA/genética , Replicação do DNA/fisiologia , Epigênese Genética/genética , Histonas/metabolismo , Humanos
5.
Toxicol Appl Pharmacol ; 329: 58-66, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28551108

RESUMO

Karenia brevis, the Florida red tide dinoflagellate produces a suite of neurotoxins known as the brevetoxins. The most abundant of the brevetoxins PbTx-2, was found to inhibit the thioredoxin-thioredoxin reductase system, whereas the PbTx-3 has no effect on this system. On the other hand, PbTx-2 activates the reduction of small disulfides such as 5,5'-dithio-bis-(2-nitrobenzoic acid) by thioredoxin reductase. PbTx-2 has an α, ß-unsaturated aldehyde moiety which functions as an efficient electrophile and selenocysteine conjugates are readily formed. PbTx-2 blocks the inhibition of TrxR by the inhibitor curcumin, whereas curcumin blocks PbTx-2 activation of TrxR. It is proposed that the mechanism of inhibition of thioredoxin reduction is via the formation of a Michael adduct between selenocysteine and the α, ß-unsaturated aldehyde moiety of PbTx-2. PbTx-2 had no effect on the rates of reactions catalyzed by related enzymes such as glutathione reductase, glutathione peroxidase or glutaredoxin.


Assuntos
Inibidores Enzimáticos/toxicidade , Toxinas Marinhas/toxicidade , Oxocinas/toxicidade , Tiorredoxina Redutase 1/antagonistas & inibidores , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Curcumina/farmacologia , Relação Dose-Resposta a Droga , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Oxirredução , Domínios Proteicos , Ratos , Selenocisteína , Tiorredoxina Redutase 1/química , Tiorredoxina Redutase 1/metabolismo , Fatores de Tempo
6.
Nucleic Acids Res ; 43(12): 5948-60, 2015 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-25990721

RESUMO

Base excision repair (BER) of an oxidized base within a trinucleotide repeat (TNR) tract can lead to TNR expansions that are associated with over 40 human neurodegenerative diseases. This occurs as a result of DNA secondary structures such as hairpins formed during repair. We have previously shown that BER in a TNR hairpin loop can lead to removal of the hairpin, attenuating or preventing TNR expansions. Here, we further provide the first evidence that AP endonuclease 1 (APE1) prevented TNR expansions via its 3'-5' exonuclease activity and stimulatory effect on DNA ligation during BER in a hairpin loop. Coordinating with flap endonuclease 1, the APE1 3'-5' exonuclease activity cleaves the annealed upstream 3'-flap of a double-flap intermediate resulting from 5'-incision of an abasic site in the hairpin loop. Furthermore, APE1 stimulated DNA ligase I to resolve a long double-flap intermediate, thereby promoting hairpin removal and preventing TNR expansions.


Assuntos
Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Expansão das Repetições de Trinucleotídeos , DNA/química , DNA/metabolismo , DNA Ligase Dependente de ATP , DNA Ligases/metabolismo , Exodesoxirribonucleases/metabolismo , Endonucleases Flap/metabolismo , Conformação de Ácido Nucleico
7.
Environ Toxicol ; 32(8): 2004-2020, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27774770

RESUMO

Arsenite has been shown to induce a variety of oxidative damage in mammalian cells. However, the mechanisms underlying cellular responses to its adverse effects remain unknown. We previously showed that the level of Nrf2, a nuclear transcription factor significantly increased in arsenite-treated human bronchial epithelial (HBE) cells suggesting that Nrf2 is involved in responding to arsenite-induced oxidative damage. To explore how Nrf2 can impact arsenite-induced oxidative damage, in this study, we examined Nrf2 activation and its regulation upon cellular arsenite exposure as well as its effects on arsenite-induced oxidative damage in HBE cells. We found that Nrf2 mRNA and protein levels were significantly increased by arsenite in a dose- and time-dependent manner. Furthermore, we showed that over-expression of Nrf2 significantly reduced the level of arsenite-induced oxidative damage in HBE cells including DNA damage, chromosomal breakage, lipid peroxidation and depletion of antioxidants. This indicates a protective role of Nrf2 against arsenite toxicity. This was further supported by the fact that activation of Nrf2 by its agonists, tertiary butylhydroquinone (t-BHQ) and sulforaphane (SFN) resulted in the same protective effects against arsenite toxicity. Moreover, we demonstrated that arsenite-induced activation of Nrf2 was mediated by the cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) signaling pathway. This is the first evidence showing that Nrf2 protects against arsenite-induced oxidative damage through the cGMP-PKG pathway. Our study suggests that activation of Nrf2 through the cGMP-PKG signaling pathway in HBE cells may be developed as a new strategy for prevention of arsenite toxicity. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 2004-2020, 2017.


Assuntos
Arsenitos/toxicidade , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Antioxidantes/metabolismo , Linhagem Celular , Humanos , Hidroquinonas/farmacologia , Isotiocianatos/farmacologia , Fator 2 Relacionado a NF-E2/agonistas , Transdução de Sinais , Sulfóxidos
8.
Nucleic Acids Res ; 42(6): 3675-91, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24423876

RESUMO

Trinucleotide repeat (TNR) expansion is responsible for numerous human neurodegenerative diseases. However, the underlying mechanisms remain unclear. Recent studies have shown that DNA base excision repair (BER) can mediate TNR expansion and deletion by removing base lesions in different locations of a TNR tract, indicating that BER can promote or prevent TNR expansion in a damage location-dependent manner. In this study, we provide the first evidence that the repair of a DNA base lesion located in the loop region of a CAG repeat hairpin can remove the hairpin, attenuating repeat expansion. We found that an 8-oxoguanine located in the loop region of CAG hairpins of varying sizes was removed by OGG1 leaving an abasic site that was subsequently 5'-incised by AP endonuclease 1, introducing a single-strand breakage in the hairpin loop. This converted the hairpin into a double-flap intermediate with a 5'- and 3'-flap that was cleaved by flap endonuclease 1 and a 3'-5' endonuclease Mus81/Eme1, resulting in complete or partial removal of the CAG hairpin. This further resulted in prevention and attenuation of repeat expansion. Our results demonstrate that TNR expansion can be prevented via BER in hairpin loops that is coupled with the removal of TNR hairpins.


Assuntos
Reparo do DNA , Expansão das Repetições de Trinucleotídeos , DNA/biossíntese , Dano ao DNA , DNA Glicosilases/metabolismo , DNA Polimerase beta/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Endodesoxirribonucleases/metabolismo , Endonucleases Flap/metabolismo , Guanina/análogos & derivados , Guanina/metabolismo , Estresse Oxidativo , Repetições de Trinucleotídeos
9.
Nucleic Acids Res ; 42(22): 13749-63, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25428354

RESUMO

5',8-cyclo-2'-deoxypurines (cdPus) are common forms of oxidized DNA lesions resulting from endogenous and environmental oxidative stress such as ionizing radiation. The lesions can only be repaired by nucleotide excision repair with a low efficiency. This results in their accumulation in the genome that leads to stalling of the replication DNA polymerases and poor lesion bypass by translesion DNA polymerases. Trinucleotide repeats (TNRs) consist of tandem repeats of Gs and As and therefore are hotspots of cdPus. In this study, we provided the first evidence that both (5'R)- and (5'S)-5',8-cyclo-2'-deoxyadenosine (cdA) in a CAG repeat tract caused CTG repeat deletion exclusively during DNA lagging strand maturation and base excision repair. We found that a cdA induced the formation of a CAG loop in the template strand, which was skipped over by DNA polymerase ß (pol ß) lesion bypass synthesis. This subsequently resulted in the formation of a long flap that was efficiently cleaved by flap endonuclease 1, thereby leading to repeat deletion. Our study indicates that accumulation of cdPus in the human genome can lead to TNR instability via a unique lesion bypass by pol ß.


Assuntos
Dano ao DNA , DNA Polimerase beta/metabolismo , Reparo do DNA , Replicação do DNA , Desoxiadenosinas/química , Repetições de Trinucleotídeos , DNA/biossíntese , DNA/química , DNA/metabolismo , Endonucleases Flap/metabolismo , Deleção de Sequência , Moldes Genéticos
10.
Mol Neurodegener ; 19(1): 26, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38504290

RESUMO

BACKGROUND: Dynamin-related protein 1 (Drp1) plays a critical role in mitochondrial dynamics. Partial inhibition of this protein is protective in experimental models of neurological disorders such as Parkinson's disease and Alzheimer's disease. The protective mechanism has been attributed primarily to improved mitochondrial function. However, the observations that Drp1 inhibition reduces protein aggregation in such neurological disorders suggest the involvement of autophagy. To investigate this potential novel protective mechanism of Drp1 inhibition, a model with impaired autophagy without mitochondrial involvement is needed. METHODS: We characterized the effects of manganese (Mn), which causes parkinsonian-like symptoms in humans, on autophagy and mitochondria by performing dose-response studies in two cell culture models (stable autophagy HeLa reporter cells and N27 rat immortalized dopamine neuronal cells). Mitochondrial function was assessed using the Seahorse Flux Analyzer. Autophagy flux was monitored by quantifying the number of autophagosomes and autolysosomes, as well as the levels of other autophagy proteins. To strengthen the in vitro data, multiple mouse models (autophagy reporter mice and mutant Drp1+/- mice and their wild-type littermates) were orally treated with a low chronic Mn regimen that was previously reported to increase α-synuclein aggregation and transmission via exosomes. RNAseq, laser captured microdissection, immunofluorescence, immunoblotting, stereological cell counting, and behavioural studies were used. RESULTS IN VITRO: data demonstrate that at low non-toxic concentrations, Mn impaired autophagy flux but not mitochondrial function and morphology. In the mouse midbrain, RNAseq data further confirmed autophagy pathways were dysregulated but not mitochondrial related genes. Additionally, Mn selectively impaired autophagy in the nigral dopamine neurons but not the nearby nigral GABA neurons. In cells with a partial Drp1-knockdown and Drp1+/- mice, Mn induced autophagic impairment was significantly prevented. Consistent with these observations, Mn increased the levels of proteinase-K resistant α-synuclein and Drp1-knockdown protected against this pathology. CONCLUSIONS: This study demonstrates that improved autophagy flux is a separate mechanism conferred by Drp1 inhibition independent of its role in mitochondrial fission. Given that impaired autophagy and mitochondrial dysfunction are two prominent features of neurodegenerative diseases, the combined protective mechanisms targeting these two pathways conferred by Drp1 inhibition make this protein an attractive therapeutic target.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Humanos , Camundongos , Ratos , alfa-Sinucleína/metabolismo , Autofagia/fisiologia , Dinaminas/genética , Dinaminas/metabolismo , Células HeLa , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Doença de Parkinson/genética
11.
bioRxiv ; 2023 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-37425803

RESUMO

Dynamin-related protein 1 (Drp1) is typically known for its role in mitochondrial fission. A partial inhibition of this protein has been reported to be protective in experimental models of neurodegenerative diseases. The protective mechanism has been attributed primarily to improved mitochondrial function. Herein, we provide evidence showing that a partial Drp1-knockout improves autophagy flux independent of mitochondria. First, we characterized in cell and animal models that at low non-toxic concentrations, manganese (Mn), which causes parkinsonian-like symptoms in humans, impaired autophagy flux but not mitochondrial function and morphology. Furthermore, nigral dopaminergic neurons were more sensitive than their neighbouring GABAergic counterparts. Second, in cells with a partial Drp1-knockdown and Drp1 +/- mice, autophagy impairment induced by Mn was significantly attenuated. This study demonstrates that autophagy is a more vulnerable target than mitochondria to Mn toxicity. Furthermore, improving autophagy flux is a separate mechanism conferred by Drp1 inhibition independent of mitochondrial fission.

12.
ACS Appl Bio Mater ; 4(2): 1632-1639, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34430802

RESUMO

Oxidative stress can damage organs, tissues, and cells through reactive oxygen species (ROS) by oxidizing DNA, proteins, and lipids, thereby resulting in diseases. However, the underlying molecular mechanisms remain to be elucidated. In this study, employing scanning ion conductance microscopy (SICM), we explored the early responses of human embryonic kidney (HEK293H) cells to oxidative DNA damage induced by potassium chromate (K2CrO4). We found that the short term (1-2 h) exposure to a low concentration (10 µM) of K2CrO4 damaged the lipid membrane of HEK293H cells, resulting in structural defects and depolarization of the cell membrane and reducing cellular secretion activity shortly after the treatment. We further demonstrated that the K2CrO4 treatment decreased the expression of the cytoskeleton protein, ß-actin, by inducing oxidative DNA damage in the exon 4 of the ß-actin gene. These results suggest that K2CrO4 caused oxidative DNA damage in cytoskeleton genes such as ß-actin and reduced their expression, thereby disrupting the organization of the cytoskeleton beneath the cell membrane and inducing cell membrane damages. Our study provides direct evidence that oxidative DNA damage disrupted human cell membrane integrity by deregulating cytoskeleton gene expression.


Assuntos
Microscopia/métodos , Estresse Oxidativo/imunologia , Humanos
13.
Wei Sheng Yan Jiu ; 39(6): 669-73, 2010 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-21351627

RESUMO

OBJECTIVE: To explore the effect of DNA polymerase beta (pol beta) expression level on genotoxicity in mouse fibroblast cell induced by hydroquinone (HQ). METHODS: pol beta wild-type cells (pol beta +/+), pol beta null cells (pol beta -/-) and pol beta overexpressed cells (pol beta oe) were applied as a model cell system. At various concentration of HQ, the cell cytotoxicities were detected by MTT assay. The DCFH-DA fluorescent probe was used to examine the cellular ROS levels. The effects of DNA damage/repair and chromosomal damage were observed by comet assay and micronuleus assay respectively. RESULTS: MTT assay showed that the doses of HQ increased, the cells viabilities were decreased. The concentrations of HQ that inhibited cell growth by 50% (IC50) in the pol beta -/- cell were more lower than those of other cells (P < 0.05). The cellular ROS level in the three kinds of cells were increased in a concentration dependent way after treated with HQ and it was more stronger in pol beta -/- cell than those in other two kinds of cells (P < 0.05). Comet assay and micronucleus assay showed that HQ induced DNA damage and increased micronucleus formation. DNA and chromosomal damage of pol beta -/- cell were more serious than those of other cells. The DNA damage repair capacities of pol beta oe cell were more stronger than pol beta +/+ and pol beta -/- cells. CONCLUSION: Cellular ROS generation could be effectively induced by HQ, leading to DNA and chromosomal damage, pol beta overexpression could help cells response to oxidative damage and protect cells from genotoxicity induced by HQ.


Assuntos
Dano ao DNA/efeitos dos fármacos , DNA Polimerase beta/metabolismo , Fibroblastos/efeitos dos fármacos , Hidroquinonas/toxicidade , Estresse Oxidativo , Animais , Células Cultivadas , Embrião de Mamíferos , Poluentes Ambientais/toxicidade , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo
14.
Wei Sheng Yan Jiu ; 39(1): 13-8, 2010 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-20364579

RESUMO

OBJECTIVE: To clarify the relationship between the expression of pol beta and DNA damage /repair induced by Benzo(a)pyrene (BaP). METHODS: pol beta wild-type cells (pol beta +/+), pol beta null cells (pol beta -/-) and wild-type pol beta overexpressed cells (pol beta oe) which had the same genetic background were studied. Firstly, RT-PCR and Western blot targeting to pol beta were carried out to measure the expression of pol beta mRNA and protein in above three kinds of cells, then MTT test and single cell gel electrophoresis (comet assay) were used to compare cell viability and DNA damage/repair of the three kinds of cells when exposed to BaP. RESULTS: There was pol beta deletion in pol beta -/- cells and the level of pol beta mRNA and protein in pol beta oe cells was twice higher than that in pol beta +/+ cells. BaP could induce DNA damage and reduce cell viability, when compared with pol beta +/+ cells, IC50 of pol beta -/- cells was remarkably lower, DNA was prone to damage and more difficult to be repaired, on the other hand, IC50 of pol beta oe cells was obviously higher and the damage effect on DNA was weaker and prone to be repaired. CONCLUSION: Pol beta played an important role in the repair of DNA damage induced by BaP, deficiency of pol beta could decrease the DNA repair capability of cells, and overexpression of pol beta could help cells response to DNA damage and protect cells from death in a certain degree.


Assuntos
Benzo(a)pireno/toxicidade , Dano ao DNA/efeitos dos fármacos , DNA Polimerase beta/metabolismo , Reparo do DNA , Fibroblastos/citologia , Animais , Células Cultivadas , DNA Polimerase beta/genética , Embrião de Mamíferos , Poluentes Ambientais/toxicidade , Fibroblastos/enzimologia , Camundongos
15.
DNA Repair (Amst) ; 93: 102912, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-33087278

RESUMO

Trinucleotide repeat (TNR) instability is the cause of over 40 human neurodegenerative diseases and certain types of cancer. TNR instability can result from DNA replication, repair, recombination, and gene transcription. Emerging evidence indicates that DNA base damage and base excision repair (BER) play an active role in regulating somatic TNR instability. These processes may potentially modulate the onset and progression of TNR-related diseases, given that TNRs are hotspots of DNA base damage that are present in mammalian cells with a high frequency. In this review, we discuss the recent advances in our understanding of the molecular mechanisms underlying BER-mediated TNR instability. We initially discuss the roles of the BER pathway and locations of DNA base lesions in TNRs and their interplay with non-B form DNA structures in governing repeat instability. We then discuss how the coordinated activities of BER enzymes can modulate a balance between the removal and addition of TNRs to regulate somatic TNR instability. We further discuss how this balance can be disrupted by the crosstalk between BER and DNA mismatch repair (MMR) machinery resulting in TNR expansion. Finally, we suggest future directions regarding BER-mediated somatic TNR instability and its association with TNR disease prevention and treatment.


Assuntos
Reparo do DNA , Expansão das Repetições de Trinucleotídeos , Animais , DNA/metabolismo , Dano ao DNA , Reparo de Erro de Pareamento de DNA , Humanos , Repetições de Trinucleotídeos
16.
Environ Pollut ; 259: 113908, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31931413

RESUMO

N6-methyladenosine (m6A), the most abundant and reversible RNA modification, plays critical a role in tumorigenesis. However, whether m6A can regulate p53, a leading antitumor protein remains poorly understood. In this study, we explored the regulatory role of m6A on p53 activation using an arsenite-transformed keratinocyte model, the HaCaT-T cell line. We created the cell line by exposing human keratinocyte HaCaT cells to 1 µM arsenite for 5 months. We found that the cells exhibited an increased m6A level along with an aberrant expression of the methyltransferases, demethylase, and readers of m6A. Moreover, the cells exhibited decreased p53 activity and reduced p53 phosphorylation, acetylation, and transactivation with a high nucleus export rate of p53. Knockdown of the m6A methyltransferase, METTL3 significantly decreased m6A level, restoring p53 activation and inhibiting cellular transformation phenotypes in the arsenite-transformed cells. Further, using both a bioinformatics analysis and experimental approaches, we demonstrated that m6A downregulated the expression of the positive p53 regulator, PRDM2, through the YTHDF2-promoted decay of PRDM2 mRNAs. We showed that m6A upregulated the expression of the negative p53 regulator, YY1 and MDM2 through YTHDF1-stimulated translation of YY1 and MDM2 mRNA. Taken together, our study revealed the novel role of m6A in mediating arsenite-induced human keratinocyte transformation by suppressing p53 activation. This study further sheds light on the mechanisms of arsenic carcinogenesis via RNA epigenetics.


Assuntos
Adenosina/análogos & derivados , Arsenitos/toxicidade , Queratinócitos/fisiologia , Adenosina/metabolismo , Arsenitos/metabolismo , Transformação Celular Neoplásica , Humanos , Metiltransferases , Proteínas de Ligação a RNA , Proteína Supressora de Tumor p53/metabolismo
17.
Sci Rep ; 10(1): 3836, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32123273

RESUMO

There is mounting evidence of androgen receptor signaling inducing genome instability and changing DNA repair capacity in prostate cancer cells. Expression of genes associated with base excision repair (BER) is increased with prostate cancer progression and correlates with poor prognosis. Poly(ADP-ribose) polymerase (PARP) and poly(ADP-ribose) glycohydrolase (PARG) are key enzymes in BER that elongate and degrade PAR polymers on target proteins. While PARP inhibitors have been tested in clinical trials and are a promising therapy for prostate cancer patients with TMPRSS2-ERG fusions and mutations in DNA repair genes, PARG inhibitors have not been evaluated. We show that PARG is a direct androgen receptor (AR) target gene. AR is recruited to the PARG locus and induces PARG expression. Androgen ablation combined with PARG inhibition synergistically reduces BER capacity in independently derived LNCaP and LAPC4 prostate cancer cell lines. A combination of PARG inhibition with androgen ablation or with the DNA damaging drug, temozolomide, significantly reduces cellular proliferation and increases DNA damage. PARG inhibition alters AR transcriptional output without changing AR protein levels. Thus, AR and PARG are engaged in reciprocal regulation suggesting that the success of androgen ablation therapy can be enhanced by PARG inhibition in prostate cancer patients.


Assuntos
Inibidores de Glicosídeo Hidrolases/farmacologia , Glicosídeo Hidrolases/antagonistas & inibidores , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicosídeo Hidrolases/metabolismo , Humanos , Masculino , Terapia de Alvo Molecular
18.
Biochimie ; 168: 241-250, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31756402

RESUMO

Prostate cancer (PCa) progression is characterized by increased expression and transcriptional activity of the androgen receptor (AR). In the advanced stages of prostate cancer, AR significantly upregulates the expression of genes involved in DNA repair. Upregulation of expression for base excision repair (BER) related genes is associated with poor patient survival. Thus, inhibition of the BER pathway may prove to be an effective therapy for prostate cancer. Using a high throughput BER capacity screening assay, we sought to identify BER inhibitors that can synergize with castration therapy. An FDA-approved drug library was screened to identify inhibitors of BER using a fluorescence-based assay suitable for HTS. A gel-based secondary assay confirmed the reduction of BER capacity by compounds identified in the primary screen. Five compounds were then selected for further testing in the independently derived, androgen-dependent prostate cancer cell lines, LNCaP and LAPC4, and in the nonmalignant prostate derived cell lines PNT1A and RWPE1. Further analysis led to the identification of a lead compound, natamycin, as an effective inhibitor of key BER enzymes DNA polymerase ß (pol ß) and DNA Ligase I (LIG I). Natamycin significantly inhibited proliferation of PCa cells in an androgen depleted environment at 1 µM concentration, however, growth inhibition did not occur with nonmalignant prostate cell lines, suggesting that BER inhibition may improve efficacy of the castration therapies.


Assuntos
Proliferação de Células/efeitos dos fármacos , DNA Ligase Dependente de ATP/antagonistas & inibidores , DNA Polimerase beta/antagonistas & inibidores , Reparo do DNA/efeitos dos fármacos , Natamicina/farmacologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Linhagem Celular Tumoral , Bases de Dados de Produtos Farmacêuticos , Humanos , Masculino
19.
Cells ; 9(1)2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31963223

RESUMO

DNA damage and base excision repair (BER) are actively involved in the modulation of DNA methylation and demethylation. However, the underlying molecular mechanisms remain unclear. In this study, we seek to understand the mechanisms by exploring the effects of oxidative DNA damage on the DNA methylation pattern of the tumor suppressor breast cancer 1 (BRCA1) gene in the human embryonic kidney (HEK) HEK293H cells. We found that oxidative DNA damage simultaneously induced DNA demethylation and generation of new methylation sites at the CpGs located at the promoter and transcribed regions of the gene ranging from -189 to +27 in human cells. We demonstrated that DNA damage-induced demethylation was mediated by nucleotide misincorporation by DNA polymerase ß (pol ß). Surprisingly, we found that the generation of new DNA methylation sites was mediated by coordination between pol ß and the de novo DNA methyltransferase, DNA methyltransferase 3b (DNMT3b), through the interaction between the two enzymes in the promoter and encoding regions of the BRCA1 gene. Our study provides the first evidence that oxidative DNA damage can cause dynamic changes in DNA methylation in the BRCA1 gene through the crosstalk between BER and de novo DNA methylation.


Assuntos
Proteína BRCA1/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Dano ao DNA , Metilação de DNA/genética , DNA Polimerase beta/metabolismo , Estresse Oxidativo , Sequência de Bases , Guanina/análogos & derivados , Guanina/metabolismo , Células HEK293 , Humanos , Modelos Biológicos , Regiões Promotoras Genéticas , Ligação Proteica , Transcrição Gênica , DNA Metiltransferase 3B
20.
Methods Mol Biol ; 1999: 87-101, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31127571

RESUMO

Trinucleotide repeat (TNR) instability (expansion and deletion) is associated with more than 42 human neurodegenerative diseases and cancer and mediated by DNA replication, repair, recombination, and gene transcription. Somatic TNR instability is involved in the progression of TNR expansion diseases and can be modulated by DNA damage repair and gene transcription. Recent studies from our group and others have shown that DNA base damage and its repair play an active role in modulating TNR instability and are responsible for somatic age-dependent CAG repeat expansion in neurons of Huntington's disease mice induced by oxidative DNA damage. However, it remains to be elucidated how DNA damage, non-B form DNA structures, and DNA repair enzymes and cofactors can coordinate to regulate somatic TNR instability. Understanding the molecular mechanisms underlying DNA damage and repair-mediated somatic TNR instability is critically important for identification of new therapeutic targets for treatment and prevention of TNR-related diseases. Here we describe the methods to study the locations and distribution of DNA base lesions and their effects on TNR instability through DNA base excision repair in in vitro reconstituted human systems.


Assuntos
Dano ao DNA , Reparo do DNA , Genômica/métodos , Expansão das Repetições de Trinucleotídeos , DNA/genética , DNA/isolamento & purificação , DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Oligonucleotídeos/genética , Oligonucleotídeos/isolamento & purificação , Oligonucleotídeos/metabolismo , Plasmídeos/genética , Reação em Cadeia da Polimerase/métodos , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa