Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 151(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38095282

RESUMO

Cajal-Retzius (CR) cells are a transient neuron type that populate the postnatal hippocampus. To understand how the persistence of CR cells influences the maturation of hippocampal circuits, we combined a specific transgenic mouse line with viral vector injection to selectively ablate CR cells from the postnatal hippocampus. We observed layer-specific changes in the dendritic complexity and spine density of CA1 pyramidal cells. In addition, transcriptomic analysis highlighted significant changes in the expression of synapse-related genes across development. Finally, we were able to identify significant changes in the expression levels of latrophilin 2, a postsynaptic guidance molecule known for its role in the entorhinal-hippocampal connectivity. These findings were supported by changes in the synaptic proteomic content in CA1 stratum lacunosum-moleculare. Our results reveal a crucial role for CR cells in the establishment of the hippocampal network.


Assuntos
Hipocampo , Proteômica , Camundongos , Animais , Hipocampo/metabolismo , Neurônios/metabolismo , Células Piramidais , Camundongos Transgênicos
2.
Nucleic Acids Res ; 50(22): 12856-12871, 2022 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-36511855

RESUMO

UV-DDB is a DNA damage recognition protein recently discovered to participate in the removal of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxoG) by stimulating multiple steps of base excision repair (BER). In this study, we examined whether UV-DDB has a wider role in BER besides oxidized bases and found it has specificity for two known DNA substrates of alkyladenine glycosylase (AAG)/N-methylpurine DNA glycosylase (MPG): 1, N6-ethenoadenine (ϵA) and hypoxanthine. Gel mobility shift assays show that UV-DDB recognizes these two lesions 4-5 times better than non-damaged DNA. Biochemical studies indicated that UV-DDB stimulated AAG activity on both substrates by 4- to 5-fold. Native gels indicated UV-DDB forms a transient complex with AAG to help facilitate release of AAG from the abasic site product. Single molecule experiments confirmed the interaction and showed that UV-DDB can act to displace AAG from abasic sites. Cells when treated with methyl methanesulfonate resulted in foci containing AAG and UV-DDB that developed over the course of several hours after treatment. While colocalization did not reach 100%, foci containing AAG and UV-DDB reached a maximum at three hours post treatment. Together these data indicate that UV-DDB plays an important role in facilitating the repair of AAG substrates.


Assuntos
DNA Glicosilases , DNA Glicosilases/metabolismo , Dano ao DNA , Reparo do DNA , DNA/genética , DNA/metabolismo
3.
Nucleic Acids Res ; 49(3): 1470-1484, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33444446

RESUMO

Alkylating drugs are among the most often used chemotherapeutics. While cancer cells frequently develop resistance to alkylation treatments, detailed understanding of mechanisms that lead to the resistance is limited. Here, by using genome-wide CRISPR-Cas9 based screen, we identify transcriptional Mediator complex subunit 13 (MED13) as a novel modulator of alkylation response. The alkylation exposure causes significant MED13 downregulation, while complete loss of MED13 results in reduced apoptosis and resistance to alkylating agents. Transcriptome analysis identified cyclin D1 (CCND1) as one of the highly overexpressed genes in MED13 knock-out (KO) cells, characterized by shorter G1 phase. MED13 is able to bind to CCND1 regulatory elements thus influencing the expression. The resistance of MED13 KO cells is directly dependent on the cyclin D1 overexpression, and its down-regulation is sufficient to re-sensitize the cells to alkylating agents. We further demonstrate the therapeutic potential of MED13-mediated response, by applying combinatory treatment with CDK8/19 inhibitor Senexin A. Importantly, the treatment with Senexin A stabilizes MED13, and in combination with alkylating agents significantly reduces viability of cancer cells. In summary, our findings identify novel alkylation stress response mechanism dependent on MED13 and cyclin D1 that can serve as basis for development of innovative therapeutic strategies.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Ciclina D1/genética , Complexo Mediador/fisiologia , Sistemas CRISPR-Cas , Linhagem Celular , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Quinases Ciclina-Dependentes/antagonistas & inibidores , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Regulação da Expressão Gênica , Humanos , Complexo Mediador/metabolismo , Regulação para Cima
4.
Blood ; 130(13): 1523-1534, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28827409

RESUMO

Endogenous DNA damage is causally associated with the functional decline and transformation of stem cells that characterize aging. DNA lesions that have escaped DNA repair can induce replication stress and genomic breaks that induce senescence and apoptosis. It is not clear how stem and proliferating cells cope with accumulating endogenous DNA lesions and how these ultimately affect the physiology of cells and tissues. Here we have addressed these questions by investigating the hematopoietic system of mice deficient for Rev1, a core factor in DNA translesion synthesis (TLS), the postreplicative bypass of damaged nucleotides. Rev1 hematopoietic stem and progenitor cells displayed compromised proliferation, and replication stress that could be rescued with an antioxidant. The additional disruption of Xpc, essential for global-genome nucleotide excision repair (ggNER) of helix-distorting nucleotide lesions, resulted in the perinatal loss of hematopoietic stem cells, progressive loss of bone marrow, and fatal aplastic anemia between 3 and 4 months of age. This was associated with replication stress, genomic breaks, DNA damage signaling, senescence, and apoptosis in bone marrow. Surprisingly, the collapse of the Rev1Xpc bone marrow was associated with progressive mitochondrial dysfunction and consequent exacerbation of oxidative stress. These data reveal that, to protect its genomic and functional integrity, the hematopoietic system critically depends on the combined activities of repair and replication of helix-distorting oxidative nucleotide lesions by ggNER and Rev1-dependent TLS, respectively. The error-prone nature of TLS may provide mechanistic understanding of the accumulation of mutations in the hematopoietic system upon aging.


Assuntos
Dano ao DNA/genética , Reparo do DNA/genética , Sistema Hematopoético/fisiologia , Estresse Oxidativo , Animais , Apoptose , Medula Óssea/patologia , Proliferação de Células , Senescência Celular/genética , DNA Polimerase Dirigida por DNA , Genoma , Células-Tronco Hematopoéticas/patologia , Camundongos , Nucleotidiltransferases
5.
Nucleic Acids Res ; 45(5): 2600-2614, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-27994034

RESUMO

Ribonucleotides (rNs) incorporated in the genome by DNA polymerases (Pols) are removed by RNase H2. Cytidine and guanosine preferentially accumulate over the other rNs. Here we show that human Pol η can incorporate cytidine monophosphate (rCMP) opposite guanine, 8-oxo-7,8-dihydroguanine, 8-methyl-2΄-deoxyguanosine and a cisplatin intrastrand guanine crosslink (cis-PtGG), while it cannot bypass a 3-methylcytidine or an abasic site with rNs as substrates. Pol η is also capable of synthesizing polyribonucleotide chains, and its activity is enhanced by its auxiliary factor DNA Pol δ interacting protein 2 (PolDIP2). Human RNase H2 removes cytidine and guanosine less efficiently than the other rNs and incorporation of rCMP opposite DNA lesions further reduces the efficiency of RNase H2. Experiments with XP-V cell extracts indicate Pol η as the major basis of rCMP incorporation opposite cis-PtGG. These results suggest that translesion synthesis by Pol η can contribute to the accumulation of rCMP in the genome, particularly opposite modified guanines.


Assuntos
Dano ao DNA , Reparo do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Ribonuclease H/metabolismo , Ribonucleotídeos/metabolismo , Linhagem Celular , Monofosfato de Citidina/metabolismo , DNA/biossíntese , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , RNA/biossíntese , Xeroderma Pigmentoso/genética
6.
Chem Res Toxicol ; 30(11): 1936-1941, 2017 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-28841305

RESUMO

In human cells, only four DNA polymerases (pols) are necessary and sufficient for the duplication of the genetic information. However, more than a dozen DNA pols are required to maintain its integrity. Such a high degree of specialization makes DNA repair pols able to cope with specific lesions or repair pathways. On the other hand, the same DNA pols can have partially overlapping roles, which could result in possible conflicts of functions, if the DNA pols are not properly regulated. DNA pol λ is a typical example of such an enzyme. It is a multifunctional enzyme, endowed with special structural and biochemical properties, which make it capable of participating in different DNA repair pathways such as base excision repair, nonhomologous end joining, and translesion synthesis. However, when mutated or deregulated, DNA pol λ can also be a source of genetic instability. Its multiple roles in DNA damage tolerance and its ability in promoting tumor progression make it also a possible target for novel anticancer approaches.


Assuntos
DNA Polimerase beta/genética , DNA Polimerase beta/metabolismo , Instabilidade Genômica , Mutagênese , Animais , Dano ao DNA , Reparo do DNA , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Estresse Oxidativo
7.
Chem Res Toxicol ; 29(9): 1493-503, 2016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27404553

RESUMO

The generation of chemical alkylating agents from nitrosation of glycine and bile acid conjugates in the gastrointestinal tract is hypothesized to initiate carcinogenesis. O(6)-carboxymethylguanine (O(6)-CMG) is a product of DNA alkylation derived from nitrosated glycine. Although the tendency of the structurally related adduct O(6)-methylguanine to code for the misincoporation of TTP during DNA replication is well-established, the impact of the presence of the O(6)-CMG adduct in a DNA template on the efficiency and fidelity of translesion DNA synthesis (TLS) by human DNA polymerases (Pols) has hitherto not been described. Herein, we characterize the ability of the four human TLS Pols η, ι, κ, and ζ and the replicative Pol δ to bypass O(6)-CMG in a prevalent mutational hot-spot for colon cancer. The results indicate that Pol η replicates past O(6)-CMG, incorporating dCMP or dAMP, whereas Pol κ incorporates dCMP only, and Pol ι incorporates primarily dTMP. Additionally, the subsequent extension step was carried out with high efficiency by TLS Pols η, κ, and ζ, while Pol ι was unable to extend from a terminal mismatch. These results provide a first basis of O(6)-CMG-promoted base misincorporation by Y- and B-family polymerases potentially leading to mutational signatures associated with colon cancer.


Assuntos
Adutos de DNA/química , Adutos de DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Guanina/análogos & derivados , Guanina/química , Adutos de DNA/toxicidade , DNA Polimerase Dirigida por DNA/química , Guanina/toxicidade , Humanos , Mutagênicos/química , Mutagênicos/metabolismo , Mutagênicos/toxicidade , Mutação , Nitrosação , Proteínas Proto-Oncogênicas p21(ras)/genética
8.
Nucleic Acids Res ; 42(1): 553-66, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24097443

RESUMO

Alkylating agents often generate 3-methylcytosine (3meC) lesions that are efficiently repaired by AlkB homologues. If AlkB homologue proteins are not functional, or the number of 3meC lesions exceeds the cellular repair capacity, the damage will persist in the genome and become substrate of DNA polymerases (Pols). Though alkylating agents are present in our environment and used in the clinics, currently nothing is known about the impact of 3meC on the accuracy and efficiency of human Pols. Here we compared the 3meC bypass properties of six human Pols belonging to the three families: B (Pol δ), X (Pols ß and λ) and Y (Pols κ, ι and η). We show that under replicative conditions 3meC impairs B-family, blocks X-family, but not Y-family Pols, in particular Pols η and ι. These Pols successfully synthesize opposite 3meC; Pol ι preferentially misincorporates dTTP and Pol η dATP. The most efficient extenders from 3meC base-paired primers are Pols κ and η. Finally, using xeroderma pigmentosum variant patient cell extracts, we provide evidence that the presence of functional Pol η is mandatory to efficiently overcome 3meC by mediating complete bypass or extension. Our data suggest that Pol η is crucial for efficient 3meC bypass.


Assuntos
Citosina/análogos & derivados , Dano ao DNA , DNA Polimerase Dirigida por DNA/metabolismo , Linhagem Celular , Citosina/metabolismo , DNA/biossíntese , Reparo do DNA , DNA Polimerase Dirigida por DNA/classificação , Humanos , DNA Polimerase iota
9.
Proc Natl Acad Sci U S A ; 110(47): 18850-5, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24191025

RESUMO

The bypass of DNA lesions by the replication fork requires a switch between the replicative DNA polymerase (Pol) and a more specialized translesion synthesis (TLS) Pol to overcome the obstacle. DNA Pol δ-interacting protein 2 (PolDIP2) has been found to physically interact with Pol η, Pol ζ, and Rev1, suggesting a possible role of PolDIP2 in the TLS reaction. However, the consequences of PolDIP2 interaction on the properties of TLS Pols remain unknown. Here, we analyzed the effects of PolDIP2 on normal and TLS by five different human specialized Pols from three families: Pol δ (family B), Pol η and Pol ι (family Y), and Pol λ and Pol ß (family X). Our results show that PolDIP2 also physically interacts with Pol λ, which is involved in the correct bypass of 8-oxo-7,8-dihydroguanine (8-oxo-G) lesions. This interaction increases both the processivity and catalytic efficiency of the error-free bypass of a 8-oxo-G lesion by both Pols η and λ, but not by Pols ß or ι. Additionally, we provide evidence that PolDIP2 stimulates Pol δ without affecting its fidelity, facilitating the switch from Pol δ to Pol λ during 8-oxo-G TLS. PolDIP2 stimulates Pols λ and η mediated bypass of other common DNA lesions, such as abasic sites and cyclobutane thymine dimers. Finally, PolDIP2 silencing increases cell sensitivity to oxidative stress and its effect is further potentiated in a Pol λ deficient background, suggesting that PolDIP2 is an important mediator for TLS.


Assuntos
Dano ao DNA/genética , DNA Polimerase beta/metabolismo , Replicação do DNA/fisiologia , Guanina/análogos & derivados , Proteínas Nucleares/metabolismo , Cromatografia por Troca Iônica , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli , Fluorescência , Guanina/metabolismo , Humanos , Imunoprecipitação , Cinética , Oligonucleotídeos/genética , RNA Interferente Pequeno/genética
10.
J Am Chem Soc ; 137(14): 4728-34, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25786104

RESUMO

Human DNA polymerase η (hPol η) contributes to anticancer drug resistance by catalyzing the replicative bypass of DNA adducts formed by the widely used chemotherapeutic agent cis-diamminedichloroplatinum (cisplatin). A chemical basis for overcoming bypass-associated resistance requires greater knowledge of how small molecules influence the hPol η-catalyzed bypass of DNA adducts. In this study, we demonstrated how synthetic nucleoside triphosphates act as hPol η substrates and characterized their influence on hPol η-mediated DNA synthesis over unmodified and platinated DNA. The single nucleotide incorporation efficiency of the altered nucleotides varied by more than 10-fold and the higher incorporation rates appeared to be attributable to the presence of an additional hydrogen bond between incoming dNTP and templating base. Finally, full-length DNA synthesis in the presence of increasing concentrations of synthetic nucleotides reduced the amount of DNA product independent of the template, representing the first example of hPol η inhibition in the presence of a platinated DNA template.


Assuntos
Adutos de DNA/biossíntese , DNA Polimerase Dirigida por DNA/metabolismo , Inibidores da Síntese de Ácido Nucleico/química , Inibidores da Síntese de Ácido Nucleico/farmacologia , Nucleotídeos/química , Nucleotídeos/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Cisplatino/química , Cisplatino/metabolismo , Adutos de DNA/química , Adutos de DNA/genética , Adutos de DNA/metabolismo , DNA Polimerase Dirigida por DNA/química , Relação Dose-Resposta a Droga , Humanos , Ligação de Hidrogênio , Cinética , Modelos Moleculares , Conformação de Ácido Nucleico , Inibidores da Síntese de Ácido Nucleico/metabolismo , Nucleotídeos/metabolismo , Conformação Proteica
11.
Virol J ; 12: 7, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25638270

RESUMO

BACKGROUND: The human polyomavirus BK expresses a 66 amino-acid peptide referred to as agnoprotein. Though mutants lacking agnoprotein are severely reduced in producing infectious virions, the exact function of this peptide remains incompletely understood. To elucidate the function of agnoprotein, we searched for novel cellular interaction partners. METHODS: Yeast-two hybrid assay was performed with agnoprotein as bait against human kidney and thymus libraries. The interaction between agnoprotein and putative partners was further examined by GST pull down, co-immunoprecipitation, and fluorescence resonance energy transfer studies. Biochemical and biological studies were performed to examine the functional implication of the interaction of agnoprotein with cellular target proteins. RESULTS: Proliferating cell nuclear antigen (PCNA), which acts as a processivity factor for DNA polymerase δ, was identified as an interaction partner. The interaction between agnoprotein and PCNA is direct and occurs also in human cells. Agnoprotein exerts an inhibitory effect on PCNA-dependent DNA synthesis in vitro and reduces cell proliferation when ectopically expressed. Overexpression of PCNA restores agnoprotein-mediated inhibition of cell proliferation. CONCLUSION: Our data suggest that PCNA is a genuine interaction partner of agnoprotein and the inhibitory effect on PCNA-dependent DNA synthesis by the agnoprotein may play a role in switching off (viral) DNA replication late in the viral replication cycle when assembly of replicated genomes and synthesized viral capsid proteins occurs.


Assuntos
Replicação do DNA , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral , Vírus BK/genética , Vírus BK/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , DNA Polimerase III/genética , DNA Polimerase III/metabolismo , Humanos , Antígeno Nuclear de Célula em Proliferação/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/genética
12.
Nucleic Acids Res ; 41(1): 229-41, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23118481

RESUMO

Human DNA polymerase (pol) λ functions in base excision repair and non-homologous end joining. We have previously shown that DNA pol λ is involved in accurate bypass of the two frequent oxidative lesions, 7,8-dihydro-8-oxoguanine and 1,2-dihydro-2-oxoadenine during the S phase. However, nothing is known so far about the relationship of DNA pol λ with the S phase DNA damage response checkpoint. Here, we show that a knockdown of DNA pol λ, but not of its close homologue DNA pol ß, results in replication fork stress and activates the S phase checkpoint, slowing S phase progression in different human cancer cell lines. We furthermore show that DNA pol λ protects cells from oxidative DNA damage and also functions in rescuing stalled replication forks. Its absence becomes lethal for a cell when a functional checkpoint is missing, suggesting a DNA synthesis deficiency. Our results provide the first evidence, to our knowledge, that DNA pol λ is required for cell cycle progression and is functionally connected to the S phase DNA damage response machinery in cancer cells.


Assuntos
DNA Polimerase beta/fisiologia , Replicação do DNA , Pontos de Checagem da Fase S do Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Quinase 1 do Ponto de Checagem , Dano ao DNA , DNA Polimerase beta/antagonistas & inibidores , DNA Polimerase beta/metabolismo , Células HeLa , Humanos , Hidroxiureia/farmacologia , Estresse Oxidativo , Proteínas Quinases/metabolismo , Interferência de RNA , Reparo de DNA por Recombinação , Estresse Fisiológico/genética
13.
Proc Natl Acad Sci U S A ; 109(2): 437-42, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22203964

RESUMO

It is of pivotal importance for genome stability that repair DNA polymerases (Pols), such as Pols λ and ß, which all exhibit considerably reduced fidelity when replicating undamaged DNA, are tightly regulated, because their misregulation could lead to mutagenesis. Recently, we found that the correct repair of the abundant and highly miscoding oxidative DNA lesion 7,8-dihydro-8-oxo-2'-deoxyguanine (8-oxo-G) is performed by an accurate repair pathway that is coordinated by the MutY glycosylase homologue (MutYH) and Pol λ in vitro and in vivo. Pol λ is phosphorylated by Cdk2/cyclinA in late S and G2 phases of the cell cycle, promoting Pol λ stability by preventing it from being targeted for proteasomal degradation by ubiquitination. However, it has remained a mystery how the levels of Pol λ are controlled, how phosphorylation promotes its stability, and how the engagement of Pol λ in active repair complexes is coordinated. Here, we show that the E3 ligase Mule mediates the degradation of Pol λ and that the control of Pol λ levels by Mule has functional consequences for the ability of mammalian cells to deal with 8-oxo-G lesions. Furthermore, we demonstrate that phosphorylation of Pol λ by Cdk2/cyclinA counteracts its Mule-mediated degradation by promoting recruitment of Pol λ to chromatin into active 8-oxo-G repair complexes through an increase in Pol λ's affinity to chromatin-bound MutYH. Finally, MutYH appears to promote the stability of Pol λ by binding it to chromatin. In contrast, Pol λ not engaged in active repair on chromatin is subject for proteasomal degradation.


Assuntos
Dano ao DNA/fisiologia , DNA Glicosilases/metabolismo , DNA Polimerase beta/metabolismo , Reparo do DNA/fisiologia , Desoxiguanosina/análogos & derivados , Ubiquitina-Proteína Ligases/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Western Blotting , Desoxiguanosina/metabolismo , Desoxiguanosina/fisiologia , Células HeLa , Humanos , Peróxido de Hidrogênio , Oligonucleotídeos/genética , Oxirredução , Fosforilação , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Supressoras de Tumor , Ubiquitinação
14.
J Biol Chem ; 288(18): 12742-52, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23511638

RESUMO

The Tim-Tipin complex plays an important role in the S phase checkpoint and replication fork stability in metazoans, but the molecular mechanism underlying its biological function is poorly understood. Here, we present evidence that the recombinant human Tim-Tipin complex (and Tim alone) markedly enhances the synthetic activity of DNA polymerase ε. In contrast, no significant effect on the synthetic ability of human DNA polymerase α and δ by Tim-Tipin was observed. Surface plasmon resonance measurements and co-immunoprecipitation experiments revealed that recombinant DNA polymerase ε directly interacts with either Tim or Tipin. In addition, the results of DNA band shift assays suggest that the Tim-Tipin complex (or Tim alone) is able to associate with DNA polymerase ε bound to a 40-/80-mer DNA ligand. Our results are discussed in view of the molecular dynamics at the human DNA replication fork.


Assuntos
Proteínas de Transporte , DNA Polimerase II , DNA , Complexos Multiproteicos , Proteínas Nucleares , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , DNA/biossíntese , DNA/química , DNA/genética , DNA Polimerase II/química , DNA Polimerase II/genética , DNA Polimerase II/metabolismo , Proteínas de Ligação a DNA , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ressonância de Plasmônio de Superfície/métodos
15.
BMC Cancer ; 14: 203, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24641873

RESUMO

BACKGROUND: Companion animals like dogs frequently develop tumors with age and similarly to human malignancies, display interpatient tumoral heterogeneity. Tumors are frequently characterized with regard to their mutation spectra, changes in gene expression or protein levels. Among others, these changes affect proteins involved in the DNA damage response (DDR), which served as a basis for the development of numerous clinically relevant cancer therapies. Even though the effects of different DNA damaging agents, as well as DDR kinetics, have been well characterized in mammalian cells in vitro, very little is so far known about the kinetics of DDR in tumor and normal tissues in vivo. DISCUSSION: Due to (i) the similarities between human and canine genomes, (ii) the course of spontaneous tumor development, as well as (iii) common exposure to environmental agents, canine tumors are potentially an excellent model to study DDR in vivo. This is further supported by the fact that dogs show approximately the same rate of tumor development with age as humans. Though similarities between human and dog osteosarcoma, as well as mammary tumors have been well established, only few studies using canine tumor samples addressed the importance of affected DDR pathways in tumor progression, thus leaving many questions unanswered. SUMMARY: Studies in humans showed that misregulated DDR pathways play an important role during tumor development, as well as in treatment response. Since dogs are proposed to be a good tumor model in many aspects of cancer research, we herein critically investigate the current knowledge of canine DDR and discuss (i) its future potential for studies on the in vivo level, as well as (ii) its possible translation to veterinary and human medicine.


Assuntos
Dano ao DNA , Reparo do DNA , Doenças do Cão/genética , Neoplasias/genética , Neoplasias/veterinária , Animais , Doenças do Cão/patologia , Cães , Evolução Molecular , Genoma , Instabilidade Genômica , Humanos , Neoplasias/patologia , Pesquisa Translacional Biomédica
16.
Nucleic Acids Res ; 40(17): 8449-59, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22753033

RESUMO

Reactive oxygen species constantly generated as by-products of cellular metabolism readily attack genomic DNA creating mutagenic lesions such as 7,8-dihydro-8-oxo-guanine (8-oxo-G) that promote aging. 8-oxo-G:A mispairs arising during DNA replication are eliminated by base excision repair initiated by the MutY DNA glycosylase homologue (MUTYH). Here, by using formaldehyde crosslinking in mammalian cell extracts, we demonstrate that the WRN helicase/exonuclease defective in the premature aging disorder Werner syndrome (WS) is recruited to DNA duplex containing an 8-oxo-G:A mispair in a manner dependent on DNA polymerase λ (Polλ) that catalyzes accurate DNA synthesis over 8-oxo-G. Similarly, by immunofluorescence, we show that Polλ is required for accumulation of WRN at sites of 8-oxo-G lesions in human cells. Moreover, we show that nuclear focus formation of WRN and Polλ induced by oxidative stress is dependent on ongoing DNA replication and on the presence of MUTYH. Cell viability assays reveal that depletion of MUTYH suppresses the hypersensitivity of cells lacking WRN and/or Polλ to oxidative stress. Biochemical studies demonstrate that WRN binds to the catalytic domain of Polλ and specifically stimulates DNA gap filling by Polλ over 8-oxo-G followed by strand displacement synthesis. Our results suggest that WRN promotes long-patch DNA repair synthesis by Polλ during MUTYH-initiated repair of 8-oxo-G:A mispairs.


Assuntos
Pareamento Incorreto de Bases , DNA Glicosilases/metabolismo , Reparo do DNA , Exodesoxirribonucleases/metabolismo , Estresse Oxidativo , RecQ Helicases/metabolismo , Animais , Linhagem Celular , DNA/metabolismo , Dano ao DNA , DNA Polimerase beta/metabolismo , Replicação do DNA , Exodesoxirribonucleases/fisiologia , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Camundongos , RecQ Helicases/fisiologia , Fase S/genética , Helicase da Síndrome de Werner
17.
DNA Repair (Amst) ; 135: 103632, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38280242

RESUMO

Neurodevelopment is a tightly coordinated process, during which the genome is exposed to spectra of endogenous agents at different stages of differentiation. Emerging evidence indicates that DNA damage is an important feature of developing brain, tightly linked to gene expression and neuronal activity. Some of the most frequent DNA damage includes changes to DNA bases, which are recognized by DNA glycosylases and repaired through base excision repair (BER) pathway. The only mammalian DNA glycosylase able to remove frequent alkylated DNA based is alkyladenine DNA glycosylase (Aag, aka Mpg). We recently demonstrated that, besides its role in DNA repair, AAG affects expression of neurodevelopmental genes in human cells. Aag was further proposed to act as reader of epigenetic marks, including 5-hydroxymethylcytosine (5hmC), in the mouse brain. Despite the potential Aag involvement in the key brain processes, the impact of Aag loss on developing brain remains unknown. Here, by using Aag knockout (Aag-/-) mice, we show that Aag absence leads to reduced DNA break levels, evident in lowered number of γH2AX foci in postnatal day 5 (P5) hippocampi. This is accompanied by changes in 5hmC signal intensity in different hippocampal regions. Transcriptome analysis of hippocampi and prefrontal cortex, at different developmental stages, indicates that lack of Aag alters gene expression, primarily of genes involved in regulation of response to stress. Across all developmental stages tested aldehyde dehydrogenase 2 (Aldh2) emerged as one of the most prominent genes deregulated in Aag-dependent manner. In line with the changes in hippocampal DNA damage levels and the gene expression, adult Aag-/- mice exhibit altered behavior, evident in decreased anxiety levels determined in the Elevated Zero Maze and increased alternations in the Elevated T Maze tests. Taken together these results suggests that Aag has functions in modulation of genome dynamics during brain development, important for animal behavior.


Assuntos
DNA Glicosilases , Humanos , Camundongos , Animais , DNA Glicosilases/genética , DNA Glicosilases/metabolismo , DNA , Ansiedade/genética , Encéfalo/metabolismo , Expressão Gênica , Mamíferos/genética
18.
Front Immunol ; 14: 1173605, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37435074

RESUMO

Human metapneumovirus (HMPV) is a pneumovirus that may cause severe respiratory disease in humans. HMPV infection has been found to increase susceptibility to bacterial superinfections leading to increased morbidity and mortality. The molecular mechanisms underlying HMPV-mediated increase in bacterial susceptibility are poorly understood and largely understudied. Type I interferons (IFNs), while critical for antiviral defenses, may often have detrimental effects by skewing the host immune response and cytokine output of immune cells. It is currently unknown if HMPV skews the inflammatory response in human macrophages triggered by bacterial stimuli. Here we report that HMPV pre-infection impacts production of specific cytokines. HMPV strongly suppresses IL-1ß transcription in response to LPS or heat-killed Pseudomonas aeruginosa and Streptococcus pneumonia, while enhancing mRNA levels of IL-6, TNF-α and IFN-ß. We demonstrate that in human macrophages the HMPV-mediated suppression of IL-1ß transcription requires TANK-binding kinase 1 (TBK1) and signaling via the IFN-ß-IFNAR axis. Interestingly, our results show that HMPV pre-infection did not impair the LPS-stimulated activation of NF-κB and HIF-1α, transcription factors that stimulate IL-1ß mRNA synthesis in human cells. Furthermore, we determined that sequential HMPV-LPS treatment resulted in accumulation of the repressive epigenetic mark H3K27me3 at the IL1B promoter. Thus, for the first time we present data revealing the molecular mechanisms by which HMPV shapes the cytokine output of human macrophages exposed to bacterial pathogens/LPS, which appears to be dependent on epigenetic reprogramming at the IL1B promoter leading to reduced synthesis of IL-1ß. These results may improve current understanding of the role of type I IFNs in respiratory disease mediated not only by HMPV, but also by other respiratory viruses that are associated with superinfections.


Assuntos
Infecções Bacterianas , Interferon beta , Interleucina-1beta , Infecções por Paramyxoviridae , Superinfecção , Humanos , Citocinas , Metapneumovirus , Transcrição Gênica , Infecções Bacterianas/imunologia , Infecções por Paramyxoviridae/imunologia
19.
Genome Biol ; 24(1): 216, 2023 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-37773136

RESUMO

BACKGROUND: Oxidation Resistance 1 (OXR1) gene is a highly conserved gene of the TLDc domain-containing family. OXR1 is involved in fundamental biological and cellular processes, including DNA damage response, antioxidant pathways, cell cycle, neuronal protection, and arginine methylation. In 2019, five patients from three families carrying four biallelic loss-of-function variants in OXR1 were reported to be associated with cerebellar atrophy. However, the impact of OXR1 on cellular functions and molecular mechanisms in the human brain is largely unknown. Notably, no human disease models are available to explore the pathological impact of OXR1 deficiency. RESULTS: We report a novel loss-of-function mutation in the TLDc domain of the human OXR1 gene, resulting in early-onset epilepsy, developmental delay, cognitive disabilities, and cerebellar atrophy. Patient lymphoblasts show impaired cell survival, proliferation, and hypersensitivity to oxidative stress. These phenotypes are rescued by TLDc domain replacement. We generate patient-derived induced pluripotent stem cells (iPSCs) revealing impaired neural differentiation along with dysregulation of genes essential for neurodevelopment. We identify that OXR1 influences histone arginine methylation by activating protein arginine methyltransferases (PRMTs), suggesting OXR1-dependent mechanisms regulating gene expression during neurodevelopment. We model the function of OXR1 in early human brain development using patient-derived brain organoids revealing that OXR1 contributes to the spatial-temporal regulation of histone arginine methylation in specific brain regions. CONCLUSIONS: This study provides new insights into pathological features and molecular underpinnings associated with OXR1 deficiency in patients.


Assuntos
Cerebelo , Histonas , Proteínas Mitocondriais , Doenças Neurodegenerativas , Humanos , Arginina/genética , Arginina/metabolismo , Atrofia , Histonas/metabolismo , Metilação , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mutação , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Cerebelo/patologia
20.
Proc Natl Acad Sci U S A ; 106(43): 18201-6, 2009 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-19820168

RESUMO

Reactive oxygen species (ROS) interact with DNA, frequently generating highly mutagenic 7,8-dihydro-8-oxoguanine (8-oxo-G) lesions. Replicative DNA polymerases (pols) often misincorporate adenine opposite 8-oxo-G. The subsequent repair mechanism allowing the removal of adenine and formation of C:8-oxo-G base pair is essential to prevent C:G to A:T transversion mutations. Here, we show by immunofluorescence experiments, in cells exposed to ROS, the involvement of MutY glycosylase homologue (MUTYH) and DNA pol lambda in the repair of A:8-oxo-G mispairs. We observe specific recruitment of MUTYH, DNA pol lambda, proliferating cell nuclear antigen (PCNA), flap endonuclease 1 (FEN1) and DNA ligases I and III from human cell extracts to A:8-oxo-G DNA, but not to undamaged DNA. Using purified human proteins and a DNA template, we reconstitute the full pathway for the faithful repair of A:8-oxo-G mispairs involving MUTYH, DNA pol lambda, FEN1, and DNA ligase I. These results reveal a cellular response pathway to ROS, important to sustain genomic stability and modulate carcinogenesis.


Assuntos
DNA Glicosilases/metabolismo , DNA Polimerase beta/metabolismo , Reparo do DNA , Guanina/análogos & derivados , Dano ao DNA , DNA Glicosilases/genética , DNA Ligase Dependente de ATP , DNA Ligases/metabolismo , DNA Polimerase beta/genética , Replicação do DNA , Instabilidade Genômica , Guanina/metabolismo , Células HeLa , Humanos , Mutagênese , Oxirredução , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA